Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
ACS Sens ; 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39388367

RESUMO

MicroRNAs (miRNAs) in dermal interstitial fluid (ISF) have recently been recognized as clinically promising biomarkers for the diagnosis and prognosis of cancer. However, the detection poses significant challenges, primarily due to the low abundance of miRNAs and the limitations of current sampling techniques. To address this issue, we develop novel porous microneedles (PMNs) array-based sensor composed of poly(vinyl alcohol) porous hydrogel and DNA-templated silver nanoclusters (AgNCs) to facilitate the enrichment and highly sensitive detection of ISF miRNA. Leveraging the capillary action facilitated by its unique porous structure and the swelling properties of the hydrogel, the PMNs array can efficiently extract 2.7 ± 0.3 mg of ISF within 5 min. Additionally, the interconnected pores within the PMNs array contribute to an increased specific surface area, thereby offering a convenient platform for the decoration of DNA-templated AgNCs. The immobilized large amount of AgNCs effectively capture the target miRNA from the extracted ISF, resulting in miRNA-induced fluorescence quenching of AgNCs. Subsequently, the introduction of the duplex-specific nuclease leads to the cleavage of DNA in DNA-RNA heteroduplexes, which release miRNA to interact with other AgNCs. This process of target recycling triggers a further reduction in fluorescence intensity, thereby enabling sensitive detection of the low-abundant miRNA down to 1.6 pM. Both in vitro and in vivo experiments validate the efficacy of the AgNCs immobilized PMNs array for the detection of miRNA biomarkers in ISF within minutes. These results indicate that the proposed PMNs array-based sensor holds great potential for the development of noninvasive personalized diagnostic strategies.

2.
Int J Biol Macromol ; 239: 124164, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37011744

RESUMO

In this study, an efficient ultrasonic-assisted extraction method was used for the extraction and optimization of four wine grape polysaccharides. A three-level, three-factor Box Behnken Design combined with the response surface approach was used to optimize the extraction conditions. Their physicochemical properties, molecular structure, antioxidant activity, immunomodulatory activity and hepatoprotective effects were examined and compared. These findings suggest that the four wine grape polysaccharides share similar basic structural features and monosaccharide composition. Furthermore, four wine grape polysaccharides exhibited antioxidant and immunomodulatory activities in a concentration-dependent manner. Moldova (MD) polysaccharide displayed better antioxidant activity and immunomodulatory ability. Furthermore, MD polysaccharide has a significant therapeutic effect on CCl4-induced rat liver injury by improving the antioxidant defense system and inhibiting oxidative stress, indicating that MD has a hepatoprotective effect. Taken together, the MD wine grape polysaccharide may have potential applications in prevention of liver disease in the functional food and pharmaceutical industries.


Assuntos
Vitis , Vinho , Ratos , Animais , Antioxidantes/farmacologia , Antioxidantes/química , Estresse Oxidativo , Polissacarídeos/farmacologia , Polissacarídeos/química
3.
Int J Pharm ; 630: 122376, 2023 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-36400133

RESUMO

High levels of proinflammatory cytokines, macrophage polarization status and immune-mediated angiogenesis play pivotal roles in the pathogenesis of inflammatory bowel disease (IBD). Thalidomide, an anti-inflammatory, immunomodulatory and antiangiogenic agent, is used off-label for treatment of IBD. The therapeutic potential of thalidomide is limited by its poor solubility and side effects associated with its systemic exposure. To address these issues and promote its therapeutic effects on IBD, thalidomide nanocrystals (Thali NCs) were prepared and coated with polydopamine (PDA), a potential macrophage polarization modulator, to form PDA coated Thali NCs (Thali@PDA). Thali@PDA possessed a high drug loading and displayed average particle size of 764.7 ± 50.30 nm. It showed a better anti-colitis effect than bare thalidomide nanocrystals at the same dose of thalidomide. Synergistic effects of polydopamine on anti-inflammatory and anti-angiogenic activities of thalidomide were observed. Furthermore, PDA coating could direct polarization of macrophages towards M2 phenotype, which boosted therapeutic effects of Thali@PDA on IBD. Upon repeated dosing of Thali@PDA for one week, symptoms of IBD in mice were significantly relieved, and histomorphology of the colitis colons were normalized. Key proinflammatory cytokine levels in the inflamed intestines were significantly decreased. Toxicity study also revealed that Thali@PDA is a safe formulation.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Nanopartículas , Camundongos , Animais , Talidomida/farmacologia , Inibidores da Angiogênese/efeitos adversos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/patologia , Anti-Inflamatórios/uso terapêutico , Doenças Inflamatórias Intestinais/tratamento farmacológico , Macrófagos , Citocinas , Sulfato de Dextrana/farmacologia
4.
Sci Rep ; 12(1): 21795, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36526664

RESUMO

Non-invasive and cost-effective diagnosis of gastric cancer is essential to improve outcomes. Aim of the study was to establish a neural network model based on patient demographic data and serum biomarker panels to aid gastric cancer diagnosis. A total of 295 patients hospitalized in Nanjing Drum Tower hospital diagnosed with gastric cancer based on tissue biopsy, and 423 healthy volunteers were included in the study. Demographical information and tumor biomarkers were obtained from Hospital Information System (HIS) as original data. Pearson's correlation analysis was applied on 574 individuals' data (training set, 229 patients and 345 healthy volunteers) to analyze the relationship between each variable and the final diagnostic result. And independent sample t test was used to detect the differences of the variables. Finally, a neural network model based on 14 relevant variables was constructed. The model was tested on the validation set (144 individuals including 66 patients and 78 healthy volunteers). The predictive ability of the proposed model was compared with other common machine learning models including logistic regression and random forest. Tumor markers contributing significantly to gastric cancer screening included CA199, CA125, AFP, and CA242 were identified, which might be considered as important inspection items for gastric cancer screening. The accuracy of the model on validation set was 86.8% and the F1-score was 85.0%, which were better than the performance of other models under the same condition. A non-invasive and low-cost artificial neural network model was developed and proved to be a valuable tool to assist gastric cancer diagnosis.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Detecção Precoce de Câncer , Antígeno Ca-125 , Redes Neurais de Computação , Biomarcadores Tumorais
5.
Molecules ; 27(19)2022 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-36235242

RESUMO

Forkhead box protein 3 (FOXP3) is an important transcription factor for regulatory T cells (Tregs) and plays an important role in their immunosuppressive function. In recent years, studies have found that FOXP3 is expressed in many kinds of tumors and plays different roles in tumors' biological behaviors, including tumor proliferation, metastasis, drug resistance, and prognosis. However, the effects of FOXP3 on tumor metastasis and its interaction with traditional Chinese medicine (TCM) remain unclear. Therefore, in this review, we focus on the effects of FOXP3 on tumor metastasis and its relationship with TCM, which can provide evidence for further research and therapy in clinical settings.


Assuntos
Fatores de Transcrição Forkhead , Neoplasias , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Medicina Tradicional Chinesa , Neoplasias/metabolismo , Linfócitos T Reguladores/metabolismo
6.
J Control Release ; 350: 841-856, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36096366

RESUMO

Melanoma is a malignant skin cancer that is prone to metastasis in the early stage and has a poor prognosis. Immunomodulatory therapy for melanoma has been a hot research topic in recent years. However, low immune cell infiltration and loss of tumor immunogenicity may occur in tumors, resulting in low response rates to immunotherapy. Thus, immunomodulatory therapy is usually used in combination with chemotherapy and radiotherapy. Development of combined therapeutic strategies with low systemic toxicity, high immune responsiveness and long-term inhibition of metastasis and recurrence of melanoma is the goal of current research. In this study, the insoluble immune adjuvant imiquimod (R837) was prepared as nanocrystals and coated with polydopamine (PDA) to form R837@PDA, which was then loaded into chitosan hydrogel (CGP) to form the drug-loaded gel system, R837@PDA@CGP (RPC), to combine immunomodulation effects, induction of immunogenic cell death (ICD) effects and immune-enhancement effects. After treatment with RPC, ICD in melanoma was induced, and the infiltration rate of cytotoxic T cells (CTLs) in melanoma was also significantly enhanced, which turned the tumor itself into an in situ vaccine and boosted the cancer-immunity cycle at the tumor site. Therefore, melanoma growth, metastasis and recurrence were notably inhibited.


Assuntos
Quitosana , Hipertermia Induzida , Melanoma , Nanopartículas , Linhagem Celular Tumoral , Humanos , Hidrogéis , Imiquimode/química , Imunoterapia/métodos , Melanoma/tratamento farmacológico , Melanoma/secundário , Nanopartículas/química
7.
J Colloid Interface Sci ; 626: 364-373, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-35797871

RESUMO

Doping Mn (II) ions into iron oxide (IO) as manganese ferrite (MnIO) has been proved to be an effective strategy to improve T1 relaxivity of IO nanoparticle in recent years; however, the high T2 relaxivity of MnIO nanoparticle hampers its T1 contrast efficiency and remains a hurdle when developing contrast agent for early and accurate diagnosis. Herein, we engineered the interfacial structure of IO nanoparticle coated with manganese ferrite shell (IO@MnIO) with tunable thicknesses. The Mn-doped shell significantly improve the T1 contrast of IO nanoparticle, especially with the thickness of ∼0.8 nm. Compared to pristine IO nanoparticle, IO@MnIO nanoparticle with thickness of ∼0.8 nm exhibits nearly 2 times higher T1 relaxivity of 9.1 mM-1s-1 at 3 T magnetic field. Moreover, exclusive engineering the interfacial structure significantly lower the T2 enhancing effect caused by doped Mn (II) ions, which further limits the impairing of increased T2 relaxivity to T1 contrast imaging. IO@MnIO nanoparticles with different shell thicknesses reveal comparable T1 relaxation rates but obvious lower T2 relaxivities and r2/r1 ratios to MnIO nanoparticles with similar sizes. The desirable T1 contrast endows IO@MnIO nanoparticle to provide sufficient signal difference between normal and tumor tissue in vivo. This work provides a detailed instance of interfacial engineering to improve IO-based T1 contrast and a new guidance for designing effective high-performance T1 contrast agent for early cancer diagnosis.


Assuntos
Meios de Contraste , Nanopartículas , Meios de Contraste/química , Compostos Férricos , Nanopartículas Magnéticas de Óxido de Ferro , Imageamento por Ressonância Magnética/métodos , Compostos de Manganês/química , Nanopartículas/química
8.
J Control Release ; 348: 911-923, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35760234

RESUMO

As we step into the post-antibiotic era, the accelerated emergence of antibiotic-resistant pathogenic bacteria poses an increasingly serious threat to public health. The formation of antibiotic-resistant biofilms further challenges currently available drugs and treatment options, calling for novel strategies for effective ablation of such biofilm with minimal concern on safety and development of resistance. Herein, we report a novel type of photodynamic nanoagent, composed of chlorin e6 (Ce6)-loaded water-soluble chitosan-coated iron oxide nanoparticles (named Ce6@WCS-IONP), for drug-resistant bacteria killing and biofilm eradication. The fabricated Ce6@WCS-IONP has negligible toxicity to mammalian cells and exhibited equivalent singlet oxygen generation capacity to free Ce6; however, its association with methicillin-resistant Staphylococcus aureus (MRSA) was greatly enhanced, as evidenced by flow cytometry analysis and transmission electron microscope. In vitro studies verified that Ce6@WCS-IONP has superior photodynamic bactericidal effect against planktonic MRSA. Furthermore, with the aid of the cationic nature and small size, Ce6@WCS-IONP could effectively penetrate into MRSA biofilm, revealed by 3D fluorescence imaging. Both biomass analysis and viable bacteria counting demonstrated that Ce6@WCS-IONP showed potent biofilm ablation efficacy, averagely 7.1 log unit lower than that in free Ce6 group upon identical light irradiation. In addition, local treatment of MRSA-infected mice with Ce6@WCS-IONP plus light irradiation resulted in significant antibacterial and wound healing effect, accompanied by good biocompatibility in vivo. Collectively, photosensitizer-loaded cationic IONP with effective biofilm penetration and photodynamic eradication potential might be a promising nano platform in fighting against antibiotic-resistant microbial pathogen and biofilm.


Assuntos
Clorofilídeos , Staphylococcus aureus Resistente à Meticilina , Nanopartículas , Fotoquimioterapia , Porfirinas , Animais , Antibacterianos/farmacologia , Biofilmes , Cátions/farmacologia , Clorofilídeos/farmacologia , Nanopartículas Magnéticas de Óxido de Ferro , Mamíferos , Camundongos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/farmacologia , Porfirinas/uso terapêutico
9.
Pharmaceutics ; 14(2)2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35213986

RESUMO

Oxygen dependence and anabatic hypoxia are the major factors responsible for the poor outcome of photodynamic therapy (PDT) against cancer. Combining of PDT and hypoxia-activatable bioreductive therapy has achieved remarkably improved antitumor efficacy compared to single PDT modality. However, controllable release and activation of prodrug and safety profiles of nanocarrier are still challenging in the combined PDT/hypoxia-triggered bioreductive therapy. Herein, we developed a near infrared (NIR) light-decomposable nanomicelle, consisting of PEGylated cypate (pCy) and mPEG-polylactic acid (mPEG2k-PLA2k) for controllable delivery of hypoxia-activated bioreductive prodrug (tirapazamine, TPZ) (designated TPZ@pCy), for combating metastatic breast cancer via hypoxia-enhanced phototherapies. TPZ@pCy was prepared by facile nanoprecipitation method, with good colloidal stability, excellent photodynamic and photothermal potency, favorable light-decomposability and subsequent release and activation of TPZ under irradiation. In vitro experiments demonstrated that TPZ@pCy could be quickly internalized by breast cancer cells, leading to remarkable synergistic tumor cell-killing potential. Additionally, metastatic breast tumor-xenografted mice with systematic administration of TPZ@pCy showed notable tumor accumulation, promoting tumor ablation and lung metastasis inhibition with negligible toxicity upon NIR light illumination. Collectively, our study demonstrates that this versatile light-decomposable polymeric micelle with simultaneous delivery of photosensitizer and bioreductive agent could inhibit tumor growth as well as lung metastasis, representing a promising strategy for potent hypoxia-enhanced phototherapies for combating metastatic breast cancer.

10.
Acta Pharm Sin B ; 11(10): 3244-3261, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34729313

RESUMO

Major challenges for cancer treatment are how to effectively eliminate primary tumor and sufficiently induce immunogenic cell death (ICD) to provoke a robust immune response for metastasis control. Here, a self-assembled cascade bioreactor was developed to improve cancer treatment with enhanced tumor penetration and synergistic therapy of starvation, chemodynamic (CDT) and photothermal therapy. Ultrasmall FeS-GOx nanodots were synthesized with glucose oxidase (GOx) as template and induced by paclitaxel (PTX) to form self-assembling FeS-GOx@PTX (FGP) via hydrophobic interaction. After accumulated at tumor sites, FGP disassembles to smaller FeS-GOx for enhanced deep tumor penetration. GOx maintains high enzymatic activity to catalyze glucose with assistant of oxygen to generate hydrogen peroxide (H2O2) as starvation therapy. Fenton reaction involving the regenerated H2O2 in turn produced more hydroxyl radicals for enhanced CDT. Following near-infrared laser at 808 nm, FGPs displayed pronounced tumor inhibition in vitro and in vivo by the combination therapy. The consequent increased exposure to calreticulin amplified ICD and promoted dendritic cells maturation. In combination with anti-CTLA4 checkpoint blockade, FGP can absolutely eliminate primary tumor and avidly inhibit distant tumors due to the enhanced intratumoral infiltration of cytotoxic T lymphocytes. Our work presents a promising strategy for primary tumor and metastasis inhibition.

11.
Int J Nanomedicine ; 16: 5167-5183, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34354350

RESUMO

INTRODUCTION: Smart theranostic nanosystems own a favorable potential to improve internalization within tumor while avoiding nonspecific interaction with normal tissues. However, development of this type of theranostic nanosystems is still a challenge. METHODS: In this study, we developed the iodine-131 (131I)-labeled multifunctional polyethylenimine (PEI)/doxorubicin (DOX) complexes with pH-controlled cellular uptake property for enhanced single-photon emission computed tomography (SPECT) imaging and chemo/radiotherapy of tumors. Alkoxyphenyl acylsulfonamide (APAS), a typical functional group that could achieve improved cellular uptake of its modified nanoparticles, was utilized to conjugate onto the functional PEI pre-modified with polyethylene glycol (PEG) with terminal groups of monomethyl ether and N-hydroxysuccinimide (mPEG-NHS), PEG with terminal groups of maleimide and succinimidyl valerate (MAL-PEG-SVA) through sulfydryl of APAS and MAL moiety of MAL-PEG-SVA. This was followed by conjugation with 3-(4'-hydroxyphenyl)propionic acid-OSu (HPAO), acetylating leftover amines of PEI, complexing DOX and labeling 131I to generate the theranostic nanosystems. RESULTS: The synthesized theranostic nanosystems exhibit favorable water solubility and stability. Every functional PEI can complex approximately 12.4 DOX, which could sustainably release of DOX following a pH-dependent manner. Remarkably, due to the surface modification of APAS, the constructed theranostic nanosystems own the capacity to achieve pH-responsive charge conversion and further lead to improved cellular uptake in cancer cells under slightly acidic condition. Above all, based on the coexistence of DOX and radioactive 131I in the single nanosystem, the synthesized nanohybrid system could afford enhanced SPECT imaging and chemo/radioactive combination therapy of cancer cells in vitro and xenografted tumor model in vivo. DISCUSSION: The developed smart nanohybrid system provides a novel strategy to improve the tumor theranostic efficiency and may be applied for different types of cancer.


Assuntos
Neoplasias , Tomografia Computadorizada de Emissão de Fóton Único , Linhagem Celular Tumoral , Quimiorradioterapia , Doxorrubicina , Humanos , Concentração de Íons de Hidrogênio , Radioisótopos do Iodo , Neoplasias/terapia , Polietilenoglicóis , Polietilenoimina
12.
J Control Release ; 335: 59-74, 2021 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-33992704

RESUMO

Novel platforms for cisplatin delivery with a controllable manner and combinable with other treatment modality to achieve synergistic antitumor effect and inhibition metastasis for treatment of triple negative breast cancer (TNBC) are highly desirable. Herein, we report a black phosphorus (BP) nanosheets-based nano-assembly which consists of cisplatin, BP, polydopamine (PDA) and hyaluronic acid (HA), cisplatin/BP/PDA-HA (CBPH), for controlled delivery of cisplatin and inhibition tumor growth as well as lung metastasis of TNBC. For constructing CBPH, the surface of BP was dual modified by PDA and HA, resulting in enhanced stability, tumor target ability and photothermal efficiency of BP. Cisplatin was released in response both to internal and external stimuli existed in tumor microenvironment, including low pH, hydrogen peroxide and NIR light, as accompanied by decomposition of BP. In vitro experiments demonstrated CBPH-treated 4 T1 cells showed elevated intracellular content of Pt and Pt-DNA adduct, which was further improved when exposure to NIR light, leading to potent antitumor effect in a synergistic pattern. Anti-metastasis studies in 2D monolayers and 3D organoids revealed that CBPH plus NIR light treatment exhibited significantly decreased migration, invasion and regrowth ability of 4 T1 cells. Furthermore, TNBC-bearing mice with systemic administrate of CBPH showed enhanced tumor accumulation of cisplatin and light-triggered inhibition of tumor growth at primary site and lung metastasis, with alleviated toxicity. But CBPH is yet to be optimized for realizing smart cisplatin delivery in response to acidic and redox stimuli in vivo. Collectively, our study demonstrates that this novel BP-based nano-assembly with controllable tumor delivery of cisplatin and metastasis inhibition of breast cancer expand the use of BP in biomedicine field and hold great promise for further development.


Assuntos
Neoplasias da Mama , Nanopartículas , Animais , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Cisplatino , Feminino , Humanos , Ácido Hialurônico , Camundongos , Fósforo , Microambiente Tumoral
13.
Angew Chem Int Ed Engl ; 60(24): 13597-13602, 2021 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-33761156

RESUMO

The iron-catalyzed alkyl-aryl coupling reaction between sulfones and arylboron compounds has remained a challenge. We report the first iron-catalyzed radical difluoroalkylation of arylborates with N-heteroaryl sulfones. The coordination between the iron catalyst and the nitrogen atom of N-heteroaryl sulfones was identified to be important in overcoming the reduction potential limitation of sulfones in the intermolecular single-electron-transfer process, which enables both fluoroalkyl N-heteroaryl sulfones (with relatively high reduction potentials) and nonfluorinated alkyl N-heteroaryl sulfones (with low reduction potentials) to serve as powerful alkylation reagents.

14.
Biomater Sci ; 9(7): 2709-2720, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33616151

RESUMO

Carbon monoxide (CO) based gas therapy has been an emerging strategy for cancer treatment. However, the uncontrolled release of CO and limited therapeutic efficacy of monotherapy are two major obstacles for clinical application. To overcome these issues, human serum albumin (HSA) nanoparticles combined with manganese dioxide (MnO2) were developed to deliver a photosensitizer (IR780) and CO donor (MnCO) for a synergistic therapy combining CO gas therapy and phototherapy. The nanoparticles (HIM-MnO2) formed catalyze hydrogen peroxide to produce oxygen for hypoxia relief. With laser irradiation, it can increase the generation of reactive oxygen species for the enhancement of photodynamic therapy (PDT). Furthermore, the generated heat of photothermal therapy (PTT) induced by nanoparticles could trigger the release of CO to achieve a therapeutic window for enhanced gas therapy. Due to the co-localization of IR780 in mitochondria, HIM-MnO2 could accumulate in mitochondria for the synergistic therapy combining CO gas therapy and phototherapy, and could oxidize the mitochondrial membrane and induce more apoptosis. After intravenous injection into tumor bearing mice, HIM-MnO2 could accumulate at tumor sites and with laser irradiation, tumor growth was significantly inhibited due to the enhanced PDT, PTT, and CO gas therapy. This study provides a strategy with oxygen generating and thermal-responsive CO release to combine phototherapy and CO gas therapy for cancer treatment.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Animais , Monóxido de Carbono , Linhagem Celular Tumoral , Compostos de Manganês/uso terapêutico , Camundongos , Mitocôndrias , Neoplasias/tratamento farmacológico , Óxidos/uso terapêutico , Oxigênio
15.
Org Lett ; 23(3): 711-715, 2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33417469

RESUMO

A novel nickel-catalyzed reductive cross-coupling between aryl iodides and difluoromethyl 2-pyridyl sulfone (2-PySO2CF2H) enables C(sp2)-C(sp2) bond formation through selective C(sp2)-S bond cleavage, which demonstrates the new reactivity of 2-PySO2CF2H reagent. This method employs readily available nickel catalyst and sulfones as cross-electrophile coupling partners, providing facile access to biaryls under mild reaction conditions without pregeneration of arylmetal reagents.

16.
Anal Chim Acta ; 1095: 138-145, 2020 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-31864614

RESUMO

Glycosylation on the cell surface contains abundant biological information, and detecting the glycan on cell surfaces can offer critical insight into biology and diseases. Here, a signal amplification strategy for the sensitive detection of glycan expression on the cell surface was proposed. In this approach, glycans on the cell surface were detected with poly(glycidyl methacrylate)-grafted silica nanosphere labeled with quantum dots (QDs) and biotin through the specific affinity reaction of avidin-biotin on the cancer cells. Glycans on the cell surface were first labeled via selective oxidization of sialyl groups into aldehydes by periodate. Aniline-catalyzed hydrazone ligation with biotin hydrazide was then used for the specific recognition to avidin. The nanoprobe was fabricated with "living" SiO2 nanoparticles with alkyl bromide groups on their surfaces. They were then subsequently grafted with poly(glycidyl methacrylate) (PGMA) brushes via the successive surface-initiated atom transfer radical polymerization. The CdTe QDs and biotin were immobilized through a ring-open reaction with epoxy groups in the PGMA brushes to obtain QDs/biotin-polymer brush-functionalized silica nanosphere (SiO2-PGMA-QDs/biotin). Enhanced sensitivity could be achieved by an increase in CdTe QDs loading per assay event, because of the large number of surface functional epoxy groups offered by the PGMA. As a result, fluorescence signal increased versus the unamplified method. This method successfully demonstrates a simple, specific, and potent method to detect glycans on the cell surface.


Assuntos
Corantes Fluorescentes/química , Nanosferas/química , Ácidos Polimetacrílicos/química , Polissacarídeos/análise , Pontos Quânticos/química , Dióxido de Silício/química , Biotina/análogos & derivados , Biotina/química , Compostos de Cádmio/química , Células HeLa , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Polissacarídeos/química , Dióxido de Silício/síntese química , Estreptavidina/química , Telúrio/química
17.
Acta Biomater ; 92: 241-253, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31078766

RESUMO

Carbon monoxide (CO)-based gas therapy has emerged as an attractive therapeutic strategy for cancer therapy. However, the main challenges are the in situ-triggered and efficient delivery of CO in tumors, which limit its further clinical application. Herein, we developed an erythrocyte membrane-biomimetic gas nanofactory (MGP@RBC) to amplify the in situ generation of CO for combined energy starvation of cancer cells and gas therapy. This nanofactory was constructed by encapsulating glucose oxidase (GOx) and Mn2(CO)10 (CO-donor) into the biocompatible polymer poly(lactic-co-glycolic acid), obtaining MGP nanoparticles, which are further covered by red blood cell (RBC) membrane. Because of the presence of proteins on RBC membranes, the nanoparticles could effectively avoid immune clearance in macrophages (Raw264.7) and significantly prolong their blood circulation time, thereby achieving higher accumulation at the tumor site. After that, the GOx in GMP@RBC could effectively catalyze the conversion of endogenous glucose to hydrogen peroxide (H2O2) in the presence of oxygen. The concomitant generation of H2O2 could efficiently trigger CO release to cause dysfunction of mitochondria and activate caspase, thereby resulting in apoptosis of the cancer cells. In addition, the depletion of intratumoral glucose could starve tumor cells by shutting down the energy supply. Altogether, the in vitro and in vivo studies of our synthesized biomimetic gas nanofactory exhibited an augmentative synergistic efficacy of CO gas therapy and energy starvation to inhibit tumor growth. It provides an attractive strategy to amplify CO generation for enhanced cancer therapy in an accurate and more efficient manner. STATEMENT OF SIGNIFICANCE: Carbon monoxide (CO) based gas therapy has emerged as an attractive therapeutic strategy for cancer therapy. In this study, we developed an erythrocyte membrane biomimetic gas nanofactory to amplify the in-situ generation of CO for combined cancer starvation and gas therapy. It is constructed by coating glucose oxidase (GOx) and CO donor-loaded nanoparticles with erythrocyte membrane. Due to the erythrocyte membrane, it can effectively prolong blood circulation time and achieve higher tumor accumulation. After accumulated in tumor, endogenous glucose can be effectively catalyzed to hydrogen peroxide, in-situ amplified CO release to induce the apoptosis of cancer cells. In addition, depleting glucose can also starve tumor cells by shutting down the energy supply. Overall, our biomimetic gas nanofactory exhibits an augmentative synergistic efficacy of CO gas therapy and starvation to increased tumor inhibition. It provide a novel strategy to deliver CO in an accurate and more efficient manner, promising for combined cancer therapy in future clinical application.


Assuntos
Biomimética , Monóxido de Carbono/uso terapêutico , Membrana Eritrocítica/química , Nanopartículas/química , Neoplasias/terapia , Animais , Antineoplásicos/farmacologia , Monóxido de Carbono/farmacocinética , Morte Celular , Linhagem Celular Tumoral , Feminino , Camundongos Endogâmicos ICR , Mitocôndrias/metabolismo , Nanopartículas/ultraestrutura , Neoplasias/patologia , Distribuição Tecidual
18.
Nanoscale ; 11(12): 5474-5488, 2019 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-30855625

RESUMO

Clinical chemotherapy for cancer is limited by the physiological barrier of tumors, resulting in low drug delivery to tumors, poor efficacy of drugs and inability to block tumor metastasis. Here we developed an intelligent switchable nitric oxide (NO)-releasing nanoparticle, IPH-NO, which loads a photosensitizer (IR780) and the chemotherapy drug paclitaxel (PTX) into NO donor-S-nitrosated human serum albumin (HSA-NO). NO exhibits two effects based on its concentration: enhancement of chemotherapy by increasing the enhanced permeability and retention (EPR) effect at low concentrations and direct killing of cancer cells at high concentrations. IPH-NO can slowly release NO in the presence of glutathione to boost tumor vascular permeability and improve drug accumulation. Near-infrared light irradiation was utilized to induce a quick release of NO that can directly kill cancer cells at high concentrations. This combination of phototherapy and NO gas therapy activated by NIR together with chemotherapy showed significant effects in tumor inhibition. Furthermore, IPH-NO blocked tumor metastasis by inhibiting epithelial mesenchymal transition. PH-NO provides a novel strategy to control NO release at tumor site for drug accumulation and combination therapies, consequently potentiating the anticancer efficacy and inhibiting tumor metastasis.


Assuntos
Nanomedicina , Nanopartículas/química , Óxido Nítrico/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glutationa/química , Meia-Vida , Humanos , Hipotermia Induzida , Raios Infravermelhos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Paclitaxel/química , Paclitaxel/farmacocinética , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacocinética , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Albumina Sérica Humana/química , Oxigênio Singlete/análise
19.
J Am Chem Soc ; 140(3): 880-883, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29278333

RESUMO

We report the first iron-catalyzed difluoromethylation of arylzincs with difluoromethyl 2-pyridyl sulfone via selective C-S bond cleavage. This method employs the readily available, bench-stable fluoroalkyl sulfone reagent and inexpensive iron catalyst, allowing facile access to structurally diverse difluoromethylated arenes at low temperatures. The experiment employing a radical clock indicates the involvement of radical species in this iron-catalyzed difluoromethylation process.

20.
Angew Chem Int Ed Engl ; 56(44): 13684-13688, 2017 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-28869355

RESUMO

Bilirubin (BR), a bile pigment that exerts potent antioxidant and anti-inflammatory effects, is also a major constituent of black pigment gallstones found in bile ducts under certain pathological conditions. Inspired by the intrinsic metal-chelating power of BR found in gallstones, herein we report a cisplatin-chelated BR-based nanoparticle (cisPt@BRNP) for use as a new photonic nanomedicine for combined photoacoustic imaging and photothermal therapy of cancers. The cisPt@BRNPs were prepared by simply mixing cisplatin with BRNPs, yielding ca. 150-nm-size NPs. Upon near-IR laser irradiation at 808 nm, cisPt@BRNPs generated considerable heat and induced clear death of cancer cells in vitro. Following intravenous injection into human colon cancer-bearing mice, cisPt@BRNPs allowed effective tumor visualization by photoacoustic imaging and remarkable antitumor efficacy by photothermal therapy, suggesting their potential for use as a new photonic nanomedicine for cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Bilirrubina/uso terapêutico , Cisplatino/uso terapêutico , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/terapia , Nanopartículas/uso terapêutico , Nanomedicina Teranóstica/métodos , Animais , Antineoplásicos/química , Bilirrubina/química , Quelantes/química , Quelantes/uso terapêutico , Cisplatino/química , Células HT29 , Humanos , Hipertermia Induzida/métodos , Raios Infravermelhos , Camundongos , Nanopartículas/química , Técnicas Fotoacústicas/métodos , Fototerapia/métodos , Platina/química , Platina/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA