Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Biosens Bioelectron ; 248: 115993, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38183788

RESUMO

Simultaneous, reliable, and ultra-sensitive analysis of promising miRNA biomarkers of colorectal cancer (CRC) in serum is critical for early diagnosis and prognosis of CRC. In this work, we proposed a novel 3D hierarchic assembly clusters-based SERS strategy with dual enrichment and enhancement designed for the ultrasensitive and quantitative analysis of two upregulated CRC-related miRNAs (miR-21 and miR-31). The biosensor contains the following: (1) SERS probe, Au nanocage@Au nanoparticles (AuNC@Au NPs) labeled with Raman reporters (RaRs). (2) magnetic capture unit, Ag-coated Fe3O4 magnetic nanoparticles (AgMNPs) modified with internal standard (IS). (3) signal amplify probes (SA probes) for the formation of hierarchic assembly clusters. Based on this sensing strategy, the intensity ratio IRaRs/IIS with Lg miRNAs presents a wide linear range (10 aM-100 pM) with a limit of detection of 3.46 aM for miR-21, 6.49 aM for miR-31, respectively. Moreover, the biosensor shows good specificity and anti-interference ability, and the reliability and repeatability of the strategy were then verified by practical detection of clinical serum. Finally, the biosensor can distinguish CRC cancer subjects from normal ones and guide the distinct tumor, lymph node, and metastasis (TNM) stages. Overall, benefiting from the face-to-face coupling of hierarchic assembly clusters, rapid magnetic enrichment and IS signal calibration of AgMNPs, the established biosensor achieves ultra-sensitive and simultaneous detection of dual miRNAs and opens potential avenues for prediction and staging of CRC.


Assuntos
Técnicas Biossensoriais , Neoplasias Colorretais , Nanopartículas Metálicas , MicroRNAs , Humanos , MicroRNAs/análise , Ouro , Reprodutibilidade dos Testes , Análise Espectral Raman , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/genética , Limite de Detecção
2.
ACS Nano ; 17(20): 20073-20086, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37792448

RESUMO

Despite the potential indicating role of tyrosinase (TYR) in cutaneous melanoma, how to capture the real changes of TYR in suspicious skin remains a major challenge. Unlike the traditional human serum test, this study reports a sensing platform that incorporates a wearable microneedle (MN) patch and trimetallic Au@Ag-Pt nanoparticles (NPs) for surface-enhanced Raman scattering (SERS) and colorimetric dual-mode detecting TYR in human skin in situ toward potential melanoma screening. In the presence of TYR, catechol immobilized on MN is preferentially oxidized to benzoquinone, which competitively impedes the interaction of MN and Au@Ag-Pt NPs, triggering the SERS-colorimetric signal reciprocal switch. Using a B16F10 mouse melanoma model, our platform is capable of noninvasively piercing the skin surface and detecting TYR levels before and during anti-PD-1 antibody treatment, which would be highly informative for prognostic judgment and illness monitoring of melanoma. Through in situ sensing for capturing the metabolic changes of TYR in advance, this platform was successfully applied to discriminate the melanoma subjects from skin moles and normal ones (p < 0.001), as well as screen potential melanoma from lactate dehydrogenase (LDH)-negative patients. Melanoma growth and prognosis can still be monitored through recording the continuous change of TYR levels. More importantly, the well-defined flexible and stretchable characteristics of the MN patch allow robustly adhering to the skin without inducing chemical or physical irritation. We believe this platform integrating MN-based in situ sensing, TYR responsiveness, and SERS/colorimetric dual-readout strategy will have high clinical importance in early diagnosis and monitoring of cutaneous melanoma.


Assuntos
Melanoma , Nanopartículas Metálicas , Neoplasias Cutâneas , Dispositivos Eletrônicos Vestíveis , Animais , Camundongos , Humanos , Melanoma/diagnóstico , Melanoma/metabolismo , Monofenol Mono-Oxigenase/metabolismo , Neoplasias Cutâneas/diagnóstico , Análise Espectral Raman , Ouro , Melanoma Maligno Cutâneo
4.
Anal Chem ; 95(35): 13101-13112, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37526338

RESUMO

A new lateral flow immunoassay strip (LFIA) combining sensitive detection and identification of multiple bacteria remains a huge challenge. In this study, we first developed multifunctional urchin-shaped Au-Ag@Pt nanoparticles (UAA@P NPs) with a unique combination of colorimetric-SERS-photothermal-catalytic (CM/SERS/PT/CL) properties and integrated them with LFIA for multiplexed detection and specific discrimination of pathogenic bacteria in blood samples. Unlike the conventional LFIA that relied on antibody (Ab), this novel LFIA introduced 4-mercaptophenylboronic acid (4-MPBA) as an ideal Ab replacer that was functionalized on UAA@P NPs (UAA@P/M NPs) with outstanding binding and enrichment capacities toward bacteria. Taking Staphylococcus aureus (S. aureus) as model bacteria, the limit of detection (LOD) was 3 CFU/mL for SERS-LFIA, 27 CFU/mL for PT-LFIA, and 18 CFU/mL for CL-LFIA, three of which were over 330-fold, 37-fold, and 55-fold more sensitive than ordinary visual CM-LFIA, respectively. Besides, this SERS-LFIA is capable of identifying three types of bacterial spiked blood samples (E. coli, S. aureus, and P. aeruginosa) effectively according to specific bacterial Raman "fingerprints" by partial least-squares-discriminant analysis (PLS-DA). More importantly, this LFIA was successfully applied to blood samples with satisfactory recoveries from 90.3% to 108.8% and capable of identifying the infected patients (N = 4) from healthy subjects (N = 2) with great accuracy. Overall, the multimodal LFIA incorporates bacteria discrimination and quantitative detection, offering an avenue for early warning and diagnosis of bacterial infection.


Assuntos
Infecções Bacterianas , Nanopartículas Metálicas , Humanos , Escherichia coli , Staphylococcus aureus , Imunoensaio , Bactérias , Anticorpos , Infecções Bacterianas/diagnóstico , Limite de Detecção , Nanopartículas Metálicas/química , Ouro/química
5.
Acta Pharm Sin B ; 13(3): 1303-1317, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36970207

RESUMO

In situ and real-time monitoring of responsive drug release is critical for the assessment of pharmacodynamics in chemotherapy. In this study, a novel pH-responsive nanosystem is proposed for real-time monitoring of drug release and chemo-phototherapy by surface-enhanced Raman spectroscopy (SERS). The Fe3O4@Au@Ag nanoparticles (NPs) deposited graphene oxide (GO) nanocomposites with a high SERS activity and stability are synthesized and labeled with a Raman reporter 4-mercaptophenylboronic acid (4-MPBA) to form SERS probes (GO-Fe3O4@Au@Ag-MPBA). Furthermore, doxorubicin (DOX) is attached to SERS probes through a pH-responsive linker boronic ester (GO-Fe3O4@Au@Ag-MPBA-DOX), accompanying the 4-MPBA signal change in SERS. After the entry into tumor, the breakage of boronic ester in the acidic environment gives rise to the release of DOX and the recovery of 4-MPBA SERS signal. Thus, the DOX dynamic release can be monitored by the real-time changes of 4-MPBA SERS spectra. Additionally, the strong T2 magnetic resonance (MR) signal and NIR photothermal transduction efficiency of the nanocomposites make it available for MR imaging and photothermal therapy (PTT). Altogether, this GO-Fe3O4@Au@Ag-MPBA-DOX can simultaneously fulfill the synergistic combination of cancer cell targeting, pH-sensitive drug release, SERS-traceable detection and MR imaging, endowing it great potential for SERS/MR imaging-guided efficient chemo-phototherapy on cancer treatment.

6.
Anal Chem ; 95(14): 5955-5966, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36916246

RESUMO

Ultra-sensitive detection of cancer-related biomarkers in serum is of great significance for early diagnosis, treatment, prognosis, and staging of cancer. In this work, we proposed a surface-enhanced Raman scattering and fluorescence (SERS/FL) dual-mode biosensor for hepatocellular carcinoma (HCC)-related miRNA (miR-224) detection using the composition of well-arranged Au nanoarrays (Au NAs) substrate coupled with the target-catalyzed hairpin assembly (CHA) strategy. The hot spots densely and uniformly distributed on the Au array offers considerably enhanced and reproducible SERS signals, along with their wide and open surface to facilitate miR-224 adsorption. By this sensing strategy, the target miR-224 can be detected in a wide linear range (1 fM to 1 nM) with a limit of detection of 0.34 fM in the SERS mode and 0.39 fM in the FL mode. Meanwhile, this biosensor with exceptional specificity and anti-interference ability can discriminate target miR-224 from other interference miRNAs. Practical analysis of human blood samples also demonstrated considerable reliability and repeatability of our developed strategy. Furthermore, this biosensor can distinguish HCC cancer subjects from normal ones and monitor HCC patients before and after hepatectomy as well as guide the distinct Barcelona clinic liver cancer (BCLC) stages. Overall, benefiting from a well-arranged Au nanoarray, CHA amplification strategy, and SERS/metal enhanced fluorescence effect, this established biosensor opens new avenues for the early prediction, warning, monitoring, and staging of HCC.


Assuntos
Técnicas Biossensoriais , Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas Metálicas , MicroRNAs , Humanos , Carcinoma Hepatocelular/diagnóstico , Ouro/química , Limite de Detecção , Neoplasias Hepáticas/diagnóstico , Nanopartículas Metálicas/química , Nanoestruturas , Reprodutibilidade dos Testes , Análise Espectral Raman , Corantes Fluorescentes/química
7.
Biosens Bioelectron ; 212: 114414, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35687957

RESUMO

Ultra-sensitive detection of pathogenic bacteria is of great significance in the early stage of bacterial infections and treatment. In this work, we report a novel strategy using multifunctional Au nano-bridged nanogap nanoparticles (Au NNPs)-based sandwich nanocomposites, that made of Concanavalin A-conjugated Fe3O4@SiO2 NPs (ConA-Fe3O4@SiO2 NPs)/bacteria/aptamer-modified Au NNPs (apt-Au NNPs), for bacteria discrimination and quantitative detection by surface-enhanced Raman scattering (SERS) and inductively coupled plasma mass spectrometry (ICP-MS), and subsequently photothermal antibacterial assay. The sandwich nanocomposite consists of ConA-Fe3O4@SiO2 NPs to magnetically enrich and photothermal killing bacteria, and dual-signal tags of apt-Au NNPs for both SERS sensing and ICP-MS quantification. This strategy can specifically distinguish different kinds of pathogenic bacteria, and provided a good linear relationship of Staphylococcus aureus (S. aureus) in the range from 50 to 104 CFU/mL with a detection limit of 11 CFU/mL, as well as realized ultralow amounts of bacterial detection in serum sample with high accuracy. Based on the quantitative detection, high antibacterial efficiency was monitored by ICP-MS. Overall, the established method combines bacteria discrimination, quantitative detection, and photothermal elimination with a simple and rapid process, which provides a novel way for the early diagnosis and treatment of bacterial infection.


Assuntos
Técnicas Biossensoriais , Nanopartículas Metálicas , Antibacterianos/farmacologia , Bactérias , Técnicas Biossensoriais/métodos , Ouro/química , Nanopartículas Metálicas/química , Dióxido de Silício/química , Análise Espectral Raman/métodos , Staphylococcus aureus
8.
Biosens Bioelectron ; 210: 114257, 2022 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-35447395

RESUMO

The simultaneous, precise, and quantitative detection of multi-components inflammatory markers (IMs) in sepsis serum by surface-enhanced Raman scattering (SERS) remains a challenging problem. A novel, multifunctional biosensor with dual enrichment and enhancement was designed for the ultrasensitive and quantitative analysis of multi-components IMs. The biosensor contains SERS tags-unique urchin core/porous shell (CPS) structure modified with Raman reporters (RaRs), magnetic assist-Ag coated Fe3O4 magnetic nanoparticles (Ag MNPs) modified with internal standard (IS), and then aptamer (Apt) modification to form the sandwich structure (Ag MNPs/IMs/CPS). This multifunctional sensor used for IMS detection has the following innovations: The intensity ratio IRaRs/IIS with Lg CIMs present a good and wide linear relationship to achieve the simultaneous, precise, and quantitative detection of IMS in serum; The detection results display ultrasensitivity, and the limit of detection (LOD) for CRP, IL-6, and PCT is 100 fg/mL, 0.1 fg/mL, and 1.0 fg/mL, which is lower than other detection techniques; The calculated data of clinical blood samples of sepsis by this SERS method is consistent with the hospital results, and can provide more compositional data of IMs. Thus, this combined approach developed a sensing platform for rapid screening, accurate evaluation, early warning, and diagnosis of sepsis.


Assuntos
Técnicas Biossensoriais , Nanopartículas Metálicas , Sepse , Técnicas Biossensoriais/métodos , Ouro/química , Humanos , Limite de Detecção , Fenômenos Magnéticos , Nanopartículas Metálicas/química , Porosidade , Sepse/diagnóstico , Análise Espectral Raman/métodos
9.
Anal Chem ; 94(15): 5785-5796, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35343684

RESUMO

New strategies combining sensitive pathogenic bacterial detection and high antimicrobial efficacy are urgently desirable. Here, we report smart triple-functional Au-Ag-stuffed nanopancakes (AAS-NPs) exhibiting (1) controllably oxidative Ag-etching thickness for simultaneously obtaining the best surface-enhanced Raman scattering (SERS) enhancement and high Ag-loading antibacterial drug delivery, (2) expressive Ag+-accelerated releasing capability under neutral phosphate-buffered saline (PBS) (pH ∼ 7.4) stimulus and robust antibacterial effectiveness involving sustainable Ag+ release, and (3) three-in-one features combining specific discrimination, sensitive detection, and inactivation of different pathogenic bacteria. Originally, AAS-NPs were synthesized by particle growth of the selective Ag-etched Au@Ag nanoparticles with K3[Fe(CN)6], followed by the formation of an unstable Prussian blue analogue for specifically binding with bacteria through the cyano group. Using specific bacterial "fingerprints" resulting from the introduction of dual-function 4-mercaptophenylboronic acid (4-MPBA, serving as both the SERS tag and internal standard) and a SERS sandwich nanostructure that was made of bacteria/SERS tags/AAS-NPs, three bacteria (E. coli, S. aureus, and P. aeruginosa) were highly sensitively discriminated and detected, with a limit of detection of 7 CFU mL-1. Meanwhile, AAS-NPs killed 99% of 1 × 105 CFU mL-1 bacteria within 60 min under PBS (pH ∼ 7.4) pretreatment. Antibacterial activities of PBS-stimulated AAS-NPs against S. aureus, E. coli, and P. aeruginosa were extraordinarily increased by 64-fold, 72-fold, and 72-fold versus PBS-untreated AAS-NPs, respectively. The multiple functions of PBS-stimulated AAS-NPs were validated by bacterial sensing, inactivation in human blood samples, and bacterial biofilm disruption. Our work exhibits an effective strategy for simultaneous bacterial sensing and inactivation.


Assuntos
Nanopartículas Metálicas , Prata , Antibacterianos/química , Antibacterianos/farmacologia , Bactérias , Escherichia coli , Ouro/química , Humanos , Nanopartículas Metálicas/química , Antígenos O , Pseudomonas aeruginosa , Prata/química , Prata/farmacologia , Análise Espectral Raman/métodos , Staphylococcus aureus
10.
Acta Biomater ; 136: 456-472, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34562660

RESUMO

The synergistic manipulation of autophagy blocking with tumor targeting and penetration effects to enhance cancer cell killing during photothermal therapy (PTT) remains a substantial challenge. Herein, we fabricated a biomimetic nanoplatform by precisely coating homologous prostate cancer cell membranes (CMs) onto the surface of mesoporous polydopamine nanoparticles (mPDA NPs) encapsulating the autophagy inhibitor chloroquine (CQ) for synergistically manipulating PTT and autophagy for anticancer treatment. The resulting biomimetic mPDA@CMs NPs-CQ system could escape macrophage phagocytosis, overcome the vascular barrier, and home in the homologous prostate tumor xenograft with high tumor targeting and penetrating efficiency. The mPDA NPs core endowed the mPDA@CMs NPs-CQ with good photothermal capability to mediate PTT killing of prostate cancer cells, while NIR-triggered CQ release from the nanosystem further arrested PTT-induced protective autophagy of cancer cells, thus weakening the resistance of prostate cancer cells to PTT. This combined PTT killing and autophagy blocking anticancer strategy could induce significant autophagosome accumulation, ROS generation, mitochondrial damage, endoplasmic reticulum stress, and apoptotic signal transduction, which finally results in synergistic prostate tumor ablation in vivo. This prostate cancer biomimetic nanosystem with synergistically enhanced anticancer efficiency achieved by manipulating PTT killing and autophagy blocking is expected to serve as a more effective anticancer strategy against prostate cancer. STATEMENT OF SIGNIFICANCE: Autophagy is considered as one of the most efficient rescuer and reinforcement mechanisms of cancer cells against photothermal therapy (PTT)-induced cancer cell eradication. How to synergistically manipulate autophagy blocking with significant tumor targeting and penetration to enhance PTT-mediated cancer cell killing remains a substantial challenge. Herein, we fabricated a biomimetic nanoplatform by precisely coating homologous cancer cell membranes onto the surface of mesoporous polydopamine nanoparticles with encapsulation of the autophagy inhibitor chloroquine for synergistic antitumor treatment with high tumor targeting and penetrating efficiency both in vitro and in vivo. This biomimetic nanosystem with synergistically enhanced anticancer efficiency by manipulating PTT killing and autophagy blocking is expected to serve as a more effective anticancer strategy.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Autofagia , Biomimética , Humanos , Indóis , Masculino , Fototerapia , Polímeros
11.
Anal Chem ; 93(25): 8799-8809, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34076420

RESUMO

Sensitive and simultaneous detection of multiple cancer-related biomarkers in serum is essential for diagnosis, therapy, prognosis, and staging of cancer. Herein, we proposed a magnetically assisted sandwich-type surface-enhanced Raman scattering (SERS)-based biosensor for ultrasensitive and multiplex detection of three hepatocellular carcinoma-related microRNA (miRNA) biomarkers. The biosensor consists of an SERS tag (probe DNA-conjugated DNA-engineered fractal gold nanoparticles, F-AuNPs) and a magnetic capture substrate (capture DNA-conjugated Ag-coated magnetic nanoparticles, AgMNPs). The proposed strategy achieved simultaneous and sensitive detection of three miRNAs (miRNA-122, miRNA-223, and miRNA-21), and the limits of detection of the three miRNAs in human serum are 349 aM for miRNA-122, 374 aM for miRNA-223, and 311 aM for miRNA-21. High selectivity and accuracy of the SERS biosensor were proved by practical analysis in human serum. Moreover, the biosensor exhibited good practicability in multiplex detection of three miRNAs in 92 clinical sera from AFP-negative patients, patients before and after hepatectomy, recurred and relapse-free patients after hepatectomy, and hepatocellular carcinoma patients at distinct Barcelona clinic liver cancer stages. The experiment results demonstrate that our SERS-based assay is a promising candidate in clinical application and exhibited potential for the prediction, diagnosis, monitoring, and staging of cancers.


Assuntos
Técnicas Biossensoriais , Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas Metálicas , MicroRNAs , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Detecção Precoce de Câncer , Fractais , Ouro , Humanos , Limite de Detecção , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , MicroRNAs/genética , Prognóstico , Análise Espectral Raman
12.
Biosens Bioelectron ; 189: 113377, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34090156

RESUMO

Monoamine oxidase inhibitors (MAOIs) are a class of drugs that can be used in the treatment of Parkinson's disease, clinical depression, and anxiety by targeting monoamine oxidase B (MAO). However, the side effects of MAOIs drive the requirement of a new framework of enzyme inhibitors development. In this context, a new type of MAOI has been built on the framework of gold nanoclusters (AuNCs), realizing the transformation from no function of small molecules to MAOI function of ligand-modified AuNCs. The MAOI activity of fabricated AuNCs can be achieved by size control and specific ligands modification. In this work, AuNCs modified with cysteamine or 4-aminothiophenol, about 1-3 nm in size, were found to have MAOI activity (MAOI-like AuNCs) and their characterization has been extensively described. Meanwhile, the possible mechanism behind this MAOI activity has been explored and it is believed that the proper size of AuNCs with ligands containing amino groups can bind tightly with the entrance to active sites of MAO, blocking the enzyme interacting with its substrates, thereby realizing the function of MAOI. Last, the antimicrobial activity and the performance of the MAOI-like AuNCs in the human blood sample were explored and suggested that MAOI-like AuNCs do not possess strong antimicrobial activity and have no visualized side effect on blood cells, although the by-product peroxide of MAO reaction may reshape the white blood cells. The research in this work may shed some light on the development of a new type of enzyme inhibitor based on the framework of nanomaterials.


Assuntos
Técnicas Biossensoriais , Preparações Farmacêuticas , Ouro , Humanos , Ligantes , Monoaminoxidase
13.
Colloids Surf B Biointerfaces ; 205: 111880, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34116399

RESUMO

Development of a multifunctional nanotherapeutic agent with high contrast-enhanced dual-modal imaging and photothermal therapy (PTT) efficacy is of great interest. Combination of ultrasound (US) and computed tomography (CT) imaging offers high spatial resolution images, showing great potential in medical imaging. Herein, the semiconducting perfluorohexane (PFH) nanodroplets, MoS2-PFH-PLLAs, are developed by stabilizing PFH droplets with the coating shell of poly (lactic-co-glycolic acid) (PLLA) and encapsulating the droplets with photoabsorbers of ultrasmall molybdenum disulfide (MoS2) nanodots. Upon near-infrared (NIR) irradiation, the MoS2-PFH-PLLAs can absorb the NIR light and convert it into heat, which not only promotes liquid-to-gas phase transition of PFH but also triggers photothermal heating, resulting in contrast-enhanced US/CT imaging and photothermal killing effect in vitro. Furthermore, the production of microbubbles can serve as the blasting agents to collaboratively enhance PTT efficacy after NIR irradiation. When intravenously injected into tumor-bearing mice, the MoS2-PFH-PLLAs exhibit a dual-modal US/CT imaging-guided synergistically therapeutic efficacy under NIR irradiation, resulting in tumor ablation. These nanotherapeutic agents demonstrate good biocompatibility, highly contrast-enhanced US/CT imaging, and combinational enhanced PTT efficacy.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Animais , Linhagem Celular Tumoral , Diagnóstico por Imagem , Fluorocarbonos , Camundongos , Molibdênio , Neoplasias/terapia , Fototerapia , Terapia Fototérmica
14.
Bioinorg Chem Appl ; 2021: 5534870, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33868396

RESUMO

How to actively target tumor sites manipulating the controllable release of the encapsulated anticancer drugs and photosensitizers for synergistic anticancer therapy remains a big challenge. In this study, a cancer cell-targeted, near-infrared (NIR) light-triggered and anticancer drug loaded liposome system (LPs) was developed for synergistic cancer therapy. Photosensitizer indocyanine green (ICG) and chemotherapy drug Curcumin (CUR) were coencapsulated into the liposomes, followed by the surface conjugation of GE11 peptide for epidermal growth factor receptor (EGFR) targeting on the cancer cell surface. Strictly controlled by NIR light, GE11 peptide modified and CUR/ICG-loaded LPs (GE11-CUR/ICG-LPs) could introduce hyperthermia in EGFR overexpressed A549 cancer cells for photothermal therapy, which could also trigger the increased release of CUR for enhanced cancer cell inhibition. GE11-CUR/ICG-LPs synergized photochemotherapy could induce reactive oxygen species (ROS) generation and cytoskeleton disruption to activate stronger apoptotic signaling events than the photothermal therapy or chemotherapy alone by regulating Bax/Bcl-2 and PI3K/AKT pathways. This EGFR-targeted drug-delivery nanosystem with NIR sensitivity may potentially serve in more effective anticancer therapeutics with reduced off-target effects.

15.
Biosens Bioelectron ; 179: 113061, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33609952

RESUMO

With the increasing number of reports on aristolochic acid I (AAI), more and more toxic and side effects have been discovered successively. The main recognized carcinogenic mechanism is that AAI is metabolized into aristololactam I (AAT) in the body by nitroreductases, ultimately forming AAT-DNA adducts that cause disease. However, the carcinogenic mechanism is still not well understood by currently reported indirect method, there has always been a great demand to develop a direct method for real-time monitoring such process. In this work, surface-enhanced Raman spectroscopy (SERS) was used for the first time to monitor the process of AAI under the action of reducing agent sodium borohydride and catalyst Raney nickel to form AAT. We first found the abundant intermediate product-amino derivative of AAI, which was never reported before by other methods. The AAT was then obtained by a one-step dehydration reaction from the amino derivative of AAI under such reduction conditions. The product of amino derivative of AAI and AAT were further verified by thin-layer chromatography, H nuclear magnetic resonance spectra, mass spectrometry, and ultra-high performance liquid chromatography. Furthermore, a density functional theory-supported in-depth vibrational characterization of AAI and AAT was performed. The monitoring of the AAI reduction process by SERS can be of great significance for further exploration of its pathogenic mechanism, prevention, and monitoring of "nephropathy" and other diseases caused by AAI.


Assuntos
Ácidos Aristolóquicos , Técnicas Biossensoriais , Nefropatias , Adutos de DNA , Humanos , Análise Espectral Raman
16.
Acta Biomater ; 121: 605-620, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33259958

RESUMO

How to enable protein degradation pathways including the autophagy-lysosome pathway (ALP) and the ubiquitin-proteasome system (UPS) to enhance the efficacy of anticancer treatments remains a substantial challenge. Cobalt oxide nanoparticles (Co3O4 NPs) have attracted interest in recent years for their potential use as a synergistic anticancer treatment, although their therapeutic mechanisms of action are still poorly understood. Here, we describe the synergistic use of Co3O4 NPs as an autophagy inhibitor, chemosensitizer and photosensitizer, which manipulate protein degradation pathways (ALP and UPS) and photothermal therapy for enhanced anticancer treatments both in vitro and in vivo. We show that Co3O4 NPs can induce autolysosome accumulation and lysosomal functions damage by inhibiting lysosomal proteolytic activity and reducing intracellular ATP levels. Notably, Co3O4 NPs can be combined with the proteasome inhibitor, Carfilzomib (Cfz), to promote the accumulation of autophagic substrates, protein ubiquitination, and endoplasmic reticulum stress, and in doing so, inhibit cancer progression. By taking advantage of their photothermal conversion efficiency, Co3O4 NPs can also serve as photothermal sensitizer, which synergistically enhances the anticancer efficacy of Cfz both in vitro and in vivo. In summary, we provide evidence of a nanomaterial-synergized, photothermal anticancer strategy that synergistically targets cancer cell survival pathways and may eventually serve to enhance the anticancer efficacy of established cancer therapeutics.


Assuntos
Nanopartículas , Fototerapia , Linhagem Celular Tumoral , Cobalto/farmacologia , Óxidos , Proteólise
17.
Anal Chem ; 92(22): 15050-15058, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33103897

RESUMO

Human monoamine oxidases (MAOs) play important roles in maintaining the homeostasis of biogenic amines. One of its isoforms, monoamine oxidase B (MAOB), is thought to be involved in several neurodegenerative diseases, which make the selective detection of MAOB activity essential. In this work, a novel surface-enhanced Raman scattering (SERS) sensor was fabricated and the MAOB activity was specifically determined by detecting the SERS signals of an enzyme-catalyzed reaction product via an amine-aldehyde click reaction. This process was simply achieved by coating core-shell gold-silver nanoparticles (Au@Ag NPs) on 3-aminopropyl aminopropyl triethoxysilane (APTES)-modified glass, and then, a monolayer of cysteamine (CA) was attached to the nanoparticle surface as a linker through Ag-S bonds. Using phenethylamine (PA) as a specific substrate of MAOB, the enzyme product phenylacetaldehyde (PAA) will produce significant Raman signals via the amine-aldehyde click reaction with CA, while other molecules, such as MAOB and PA, have no signal output because they cannot form close interaction with nanoparticles due to the existence of a CA layer. This sensor was further used for the specific determination of MAOB activity in clinical blood samples and the MAOB inhibitor assessment successfully. Meanwhile, by changing the click reaction types and taking advantage of the SERS fingerprint peaks for the specific click reaction products, this strategy offers huge potential to detect multiple enzyme activities simultaneously and can be used for new click reaction screening, enzyme-related disease diagnosis, drug screening, and clinical diagnosis.


Assuntos
Monoaminoxidase/metabolismo , Análise Espectral Raman/métodos , Aldeídos/química , Aminas/química , Química Click , Cisteamina/química , Ouro/química , Humanos , Nanopartículas Metálicas/química , Prata/química , Propriedades de Superfície
18.
Angew Chem Int Ed Engl ; 59(8): 3226-3234, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-31756258

RESUMO

Pathogenesis hallmarks for tuberculosis (TB) are the Mycobacterium tuberculosis (Mtb) escape from phagolysosomal destruction and limited drug delivery into infected cells. Several nanomaterials can be entrapped in lysosomes, but the development of functional nanomaterials to promote phagolysosomal Mtb clearance remains a big challenge. Here, we report on the bactericidal effects of selenium nanoparticles (Se NPs) against Mtb and further introduce a novel nanomaterial-assisted anti-TB strategy manipulating Ison@Man-Se NPs for synergistic drug-induced and phagolysosomal destruction of Mtb. Ison@Man-Se NPs preferentially entered macrophages and accumulated in lysosomes releasing Isoniazid. Surprisingly, Ison@Man-Se/Man-Se NPs further promoted the fusion of Mtb into lysosomes for synergistic lysosomal and Isoniazid destruction of Mtb. Concurrently, Ison@Man-Se/Man-Se NPs also induced autophagy sequestration of Mtb, evolving into lysosome-associated autophagosomal Mtb degradation linked to ROS-mitochondrial and PI3K/Akt/mTOR signaling pathways. This novel nanomaterial-assisted anti-TB strategy manipulating antimicrobial immunity and Mtb clearance may potentially serve in more effective therapeutics against TB and drug-resistant TB.


Assuntos
Antibacterianos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Isoniazida/química , Macrófagos/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Nanopartículas/química , Selênio/química , Tuberculose/tratamento farmacológico , Humanos , Tuberculose/patologia
19.
Mikrochim Acta ; 186(8): 572, 2019 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-31342193

RESUMO

A nanohybrid probe was fabricated from manganese dioxide nanosheets (MnO2 NSs), molybdenum disulfide quantum dots (MoS2 QDs) and o-phenylenediamine (OPD) for ratiometric detection of glutathione (GSH) in aqueous solutions and living cells. The MoS2 QDs act as the fluorescent "turn off-on" units. The MnO2 NSs have 3 functions, viz. (a) as fluorescence quencher, (b) as fluorescence initiator for oxidized OPD (ox OPD) and (c) as selective recognizer of GSH. The quenched blue fluorescence of the MoS2 QDs can be restored by introducing GSH that reduces the MnO2 NSs. However, the green fluorescence of ox OPD is decreased through the loss of peroxidase activity of MnO2 NSs in the presence of GSH. Therefore, the ratio of the fluorescence intensities at 560 and 400 nm (from ox OPD and MoS2 QDs, respectively) linearly decreases with increasing concentrations of GSH. Under the optimal conditions, the detection limit for GSH is as low as 90 nM. The method was successfully applied to the determination of GSH in human serum samples. This nanohybrid also is shown to be membrane-permeable and to have low cytotoxicity. This paved the way to intracellular sensing of GSH in living normal HFF and cancerous HeLa cells. Additionally, by combining with logic gate, this assay was successfully applied to visually discriminate changes in the intracellular GSH. The combination of ratiometric fluorometry and peroxidase mimicking can provide a wide range of application in bioanalysis and intracellular imaging. Graphical abstract Schematic representation of the ratiometric fluorometric detection and cellular imaging of glutathione using a nanohybrid composed of MoS2 quantum dots and MnO2 nanosheets with dual (blue and green emission and peroxidase mimicking properties.


Assuntos
Glutationa/sangue , Nanoestruturas/química , Imagem Óptica/métodos , Pontos Quânticos/química , Linhagem Celular , Linhagem Celular Tumoral , Dissulfetos , Fluorometria/métodos , Glutationa/análise , Células HeLa , Humanos , Limite de Detecção , Compostos de Manganês , Molibdênio , Óxidos , Peroxidase
20.
J Cell Biochem ; 120(10): 16543-16552, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31099025

RESUMO

The integration of ultrasound (US) contrast enhancement with oxygen-loading nanoagents provide the synergistic strategy for simultaneously US imaging and hypoxic microenvironment modulation. Herein, we synthesize pentafluorobutane (PFB)-loading methoxy poly(ethylene glycol)-b-poly(l-lactide) (PLLA) nanoparticle as the novel US-contrast-enhanced agent and demonstrate that PFB@PLLA effectively loads oxygen. We characterize the nanosize, phase-transformation property and oxygen-loading amount of PFB@PLLA and investigate the effectiveness of these nanoagents in US-contrast-enhanced imaging. The PFB@PLLA displays a perfect temperature-responsive phase-transition property and its liquid-to-gas phase transition temperature is 45°C, which produces microbubbles in the targeted regions. Moreover, PFB@PLLA loads high amount of oxygen and US-triggering PFB@PLLA reoxygenation effectively inhibits the expression of hypoxia-related proteins (HIF-1α and CAIX), reduces lactate secretion and glycolysis, which modulates hypoxic microenvironment and inhibits cancer cell migration and invasion in vitro. This study demonstrates that the US contrast-enhanced activity of PFB@PLLAs and the promising utility of oxygen-loading nanoagents to improve hypoxic microenvironment.


Assuntos
Materiais Revestidos Biocompatíveis , Meios de Contraste , Hidrocarbonetos Fluorados , Nanopartículas/química , Hipóxia Celular , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Meios de Contraste/química , Meios de Contraste/farmacologia , Células Hep G2 , Humanos , Hidrocarbonetos Fluorados/química , Hidrocarbonetos Fluorados/farmacologia , Células MCF-7 , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA