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











Base de dados
Intervalo de ano de publicação
1.
Angew Chem Int Ed Engl ; 63(38): e202407037, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-38767062

RESUMO

The stimulator of interferon genes (STING) pathway is a potent therapeutic target for innate immunity. Despite the efforts to develop pocket-dependent small-molecule STING agonists that mimic the endogenous STING ligand, cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), most of these agonists showed disappointing results in clinical trials owing to the limitations of the STING pocket. In this study, we developed novel pocket-independent STING-activating agonists (piSTINGs), which act through multivalency-driven oligomerization to activate STING. Additionally, a piSTING-adjuvanted vaccine elicited a significant antibody response and inhibited tumour growth in therapeutic models. Moreover, a piSTING-based vaccine combination with aPD-1 showed remarkable potential to enhance the effectiveness of immune checkpoint blockade (ICB) immunotherapy. In particular, piSTING can strengthen the impact of STING pathway in immunotherapy and accelerate the clinical translation of STING agonists.


Assuntos
Proteínas de Membrana , Proteínas de Membrana/agonistas , Proteínas de Membrana/metabolismo , Humanos , Animais , Camundongos , Imunoterapia , Nucleotídeos Cíclicos/química , Nucleotídeos Cíclicos/farmacologia
2.
Adv Mater ; 36(16): e2308155, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38295870

RESUMO

Following the success of the dendritic cell (DC) vaccine, the cell-based tumor vaccine shows its promise as a vaccination strategy. Except for DC cells, targeting other immune cells, especially myeloid cells, is expected to address currently unmet clinical needs (e.g., tumor types, safety issues such as cytokine storms, and therapeutic benefits). Here, it is shown that an in situ injected macroporous myeloid cell adoptive scaffold (MAS) not only actively delivers antigens (Ags) that are triggered by scaffold-infiltrating cell surface thiol groups but also releases granulocyte-macrophage colony-stimulating factor and other adjuvant combos. Consequently, this promotes cell differentiation, activation, and migration from the produced monocyte and DC vaccines (MASVax) to stimulate antitumor T-cell immunity. Neoantigen-based MASVax combined with immune checkpoint blockade induces rejection of established tumors and long-term immune protection. The combined depletion of immunosuppressive myeloid cells further enhances the efficacy of MASVax, indicating the potential of myeloid cell-based therapies for immune enhancement and normalization treatment of cancer.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Vacinação , Engenharia Celular , Células Mieloides , Células Dendríticas
3.
J Med Chem ; 66(18): 13266-13279, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37676021

RESUMO

Protein or peptide cancer vaccines usually include immune potentiators, so-called adjuvants. However, it remains challenging to identify structurally simple, chemically accessible synthetic molecules that are effective and safe as vaccine adjuvant. Here, we present cholicamideß (6), a self-assembling small-molecule vaccine adjuvant with an improved toxicity profile and proven efficacy in vivo. We demonstrate that cholicamideß (6), which is less cytotoxic than its parent compound, forms virus-like particles to potently activate dendritic cells with the concomitant secretion of cytokines. When combined with a peptide antigen, cholicamideß (6) potentiated the antigen presentation on dendritic cells to induce antigen-specific T cells. As a therapeutic cancer vaccine adjuvant in mice, a mixture of cholicamideß (6) and a peptide antigen protected mice from the challenges of malignant cancer cells without overt toxicity. Cholicamideß (6) may offer a translational opportunity as an unprecedented class of small-molecule cancer vaccine adjuvants.


Assuntos
Vacinas Anticâncer , Neoplasias , Animais , Camundongos , Vacinas Anticâncer/uso terapêutico , Adjuvantes de Vacinas , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/química , Linfócitos T , Adjuvantes Farmacêuticos , Vacinas de Subunidades Antigênicas , Peptídeos , Células Dendríticas
4.
Appl Microbiol Biotechnol ; 107(12): 3967-3981, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37178306

RESUMO

α-L-Arabinofuranosidases (Abfs) play a crucial role in the degradation of hemicelluloses, especially arabinoxylans (AX). Most of the available characterized Abfs are from bacteria, while fungi, as natural decomposers, contain Abfs with little attention given. An arabinofuranosidase (ThAbf1), belonging to the glycoside hydrolase 51 (GH51) family, from the genome of the white-rot fungus Trametes hirsuta, was recombinantly expressed, characterized, and functionally determined. The general biochemical properties showed that the optimal conditions for ThAbf1 were pH 6.0 and 50°C. In substrate kinetics assays, ThAbf1 preferred small fragment arabinoxylo-oligosaccharides (AXOS) and could surprisingly hydrolyze di-substituted 23,33-di-L-arabinofuranosyl-xylotriose (A2,3XX). It also synergized with commercial xylanase (XYL) and increased the saccharification efficiency of arabinoxylan. The crystal structure of ThAbf1 indicated the presence of an adjacent cavity next to the catalytic pocket which led to the ability of ThAbf1 to degrade di-substituted AXOS. The narrow binding pocket prevents ThAbf1 from binding larger substrates. These findings have strengthened our understanding of the catalytic mechanism of GH51 family Abfs and provided a theoretical foundation for the development of more efficient and versatile Abfs to accelerate the degradation and biotransformation of hemicellulose in biomass. KEY POINTS: • ThAbf1 from Trametes hirsuta degraded di-substituted arabinoxylo-oligosaccharide. • ThAbf1 performed detailed biochemical characterization and kinetics. • ThAbf1 structure has been obtained to illustrate the substrate specificity.


Assuntos
Polyporaceae , Trametes , Xilanos/metabolismo , Polyporaceae/metabolismo , Oligossacarídeos/metabolismo , Glicosídeo Hidrolases/metabolismo , Especificidade por Substrato
6.
Nano Res ; 15(7): 6328-6339, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35464625

RESUMO

Immunostimulatory therapies based on pattern recognition receptors (PRRs) have emerged as an effective approach in the fight against cancer, with the ability to recruit tumor-specific lymphocytes in a low-immunogenicity tumor environment. The agonist cyclic dinucleotides (CDNs) of the stimulator of interferon gene (STING) are a group of very promising anticancer molecules that increase tumor immunogenicity by activating innate immunity. However, the tumor immune efficacy of CDNs is limited by several factors, including relatively narrow cytokine production, inefficient delivery to STING, and rapid clearance. In addition, a single adjuvant molecule is unable to elicit a broad cytokine response and thus cannot further amplify the anticancer effect. To address this problem, two or more agonist molecules are often used together to synergistically enhance immune efficacy. In this work, we found that a combination of the STING agonist CDGSF and the Toll-like receptor 7/8 (TLR7/8) agonist 522 produced a broader cytokine response. Subsequently, we developed multicomponent nanovaccines (MCNVs) consisting of a PC7A polymer as a nanocarrier encapsulating the antigen OVA and adjuvant molecules. These MCNVs activate bone marrow-derived dendritic cells (BMDCs) to produce multiple proinflammatory factors that promote antigen cross-presentation to stimulate specific antitumor T-cell responses. In in vivo experiments, we observed that MCNVs triggered a strong T-cell response in tumor-infiltrating lymphocytes, resulting in significant tumor regression and, notably, a 100% survival rate in mice through 25 days without other partnering therapies. These data suggest that our nanovaccines have great potential to advance cancer immunotherapy with increased durability and potency. Electronic Supplementary Material: Supplementary material (synthesis of CDGSF, 522, PC7A and OVA; preparation of MCNVs; representative gating strategies for flow cytometry) is available in the online version of this article at 10.1007/s12274-022-4282-x.

7.
BMC Med Genomics ; 15(1): 53, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35260168

RESUMO

BACKGROUND: N6-methyladenosine (m6A) RNA regulation was recently reported to be important in carcinogenesis and cancer development. However, the characteristics of m6A modification and its correlations with clinical features, genome instability, tumor microenvironments (TMEs), and immunotherapy responses in hepatocellular carcinoma (HCC) have not been fully explored. METHODS: We systematically analyzed the m6A regulator-based expression patterns of 486 patients with HCC from The Cancer Genome Atlas and Gene Expression Omnibus databases, and correlated these patterns with clinical outcomes, somatic mutations, TME cell infiltration, and immunotherapy responses. The m6A score was developed by principal component analysis to evaluate m6A modifications in individual patients. RESULTS: M6A regulators were dysregulated in HCC samples, among which 18 m6A regulators were identified as risk factors for prognosis. Three m6A regulator-based expression patterns, namely m6A clusters, were determined among HCC patients by m6A regulators with different m6A scores, somatic mutation counts, and specific TME features. Additionally, three distinct m6A regulator-associated gene-based expression patterns were also identified based on prognosis-associated genes that were differentially expressed among the three m6A clusters, showing similar properties as the m6A regulator-based expression patterns. Higher m6A scores were correlated with older age, advanced stages, lower overall survival, higher somatic mutation counts, elevated PD-L1 expression levels, and poorer responses to immune checkpoint inhibitors. The m6A score was validated as an independent and valuable prognostic factor for HCC. CONCLUSION: M6A modification is correlated with genome instability and TME in HCC. Evaluating m6A regulator-based expression patterns and the m6A score of individual tumors may help identify candidate biomarkers for prognosis prediction and immunotherapeutic strategy selection.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/genética , Instabilidade Genômica , Humanos , Neoplasias Hepáticas/genética , Metilação , Prognóstico , Microambiente Tumoral/genética
8.
Biomaterials ; 273: 120788, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33933912

RESUMO

Intrinsic immune behaviors of nanomaterials and immune systems promote research on their adjuvanticity and the design of next generation nanovaccine-based immunotherapies. Herein, we report a promising multifunctional nanoadjuvant by exploring the immune-potentiating effects of black phosphorus nanosheets (BPs) in vitro and in vivo. The facile coating of BPs with phenylalanine-lysine-phenylalanine (FKF) tripeptide-modified antigen epitopes (FKF-OVAp@BP) enables the generation of a minimalized nanovaccine by integrating high loading capacity, efficient drug delivery, comprehensive dendritic cell (DC) activation, and biocompatibility for cancer immunotherapy. Systemic immunization elicits potent antitumor cellular immunity and significantly augments checkpoint blockade (CPB) against melanoma in a mouse model. Furthermore, near-infrared (NIR) photothermal effects of BPs create an immune-favorable microenvironment for improved local immunization. This study offers new insight into the integration of immunoactivity and photothermal effects for enhanced cancer immunotherapy by using a nanoadjuvant and thus potentially advances the design and application of multifunctional adjuvant materials for cancer nanotreatment.


Assuntos
Imunoterapia , Fósforo , Adjuvantes Imunológicos , Animais , Sistemas de Liberação de Medicamentos , Fatores Imunológicos , Camundongos
9.
Chem Commun (Camb) ; 57(4): 504-507, 2021 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-33331360

RESUMO

A novel STING agonist, CDGSF, ipsilaterally modified with phosphorothioate and fluorine, was synthesized. The phosphorothioate in CDGSF might be a site for covalent conjugation. Injection of CDGSF generated an immunogenic ("hot") tumor microenvironment to suppress melanoma, more efficiently than dithio CDG. In particular, immunization with SARS-CoV-2 spike protein using CDGSF as an adjuvant elicited an exceptionally high antibody titer and a robust T cell response, overcoming the drawbacks of aluminum hydroxide. These results highlighted the therapeutic potential of CDGSF for cancer immunotherapy and the adjuvant potential of the STING agonist in the SARS-CoV-2 vaccine for the first time.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra COVID-19/administração & dosagem , COVID-19/prevenção & controle , Melanoma Experimental/tratamento farmacológico , Proteínas de Membrana/agonistas , Nucleotídeos Cíclicos/administração & dosagem , Neoplasias Cutâneas/tratamento farmacológico , Adjuvantes Imunológicos/síntese química , Hidróxido de Alumínio/administração & dosagem , Hidróxido de Alumínio/química , Animais , Anticorpos Antivirais/biossíntese , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/virologia , COVID-19/imunologia , COVID-19/virologia , Vacinas contra COVID-19/química , ELISPOT , Humanos , Imunoterapia/métodos , Interferon gama/biossíntese , Melanoma Experimental/imunologia , Melanoma Experimental/mortalidade , Melanoma Experimental/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Nucleotídeos Cíclicos/síntese química , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia , Glicoproteína da Espícula de Coronavírus/administração & dosagem , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/imunologia , Análise de Sobrevida , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/virologia , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Vacinação/métodos
10.
Biochem Biophys Res Commun ; 525(2): 360-365, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32089266

RESUMO

Aldehyde dehydrogenase 2, a mitochondrial matrix enzyme, plays a crucial role in protecting the heart against stress, such as ischemia reperfusion and alcohol injury. The present study aimed to investigate the effect of aldehyde dehydrogenase 2 on lipotoxic cardiomyopathy and to explore the possible mechanisms in vitro. Primary cardiomyocytes in the lipotoxic group were treated with oxidatively modified low-density lipoprotein (50 mg/L) for 24 h. Overexpression of aldehyde dehydrogenase 2 was achieved using the aldehyde dehydrogenase 2 activator, Alda-1 (20 µM). We found that cardiomyocyte apoptosis was attenuated by aldehyde dehydrogenase 2 overexpression. In addition, aldehyde dehydrogenase 2 overexpression inhibited the expression of BCL2 associated X, apoptosis regulator (BAX) and caspase 3, while it enhanced protein kinase B (AKT) and glycogen synthase kinase 3 beta (GSK-3ß) phosphorylation. The results suggested that aldehyde dehydrogenase 2 is cardioprotective against lipotoxic cardiomyopathy, probably by reducing apoptosis through the AKT/glycogen synthase kinase 3 beta (GSK-3ß) pathway. Our findings partially revealed the molecular mechanism of aldehyde dehydrogenase 2's cardioprotective effect against lipotoxic injury, and suggest a new therapeutic strategy to treat lipotoxic cardiomyopathy.


Assuntos
Aldeído-Desidrogenase Mitocondrial/fisiologia , Apoptose/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Cardiomiopatias/etiologia , Cardiomiopatias/terapia , Células Cultivadas , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Lipídeos/toxicidade , Lipoproteínas IDL/toxicidade , Substâncias Protetoras/farmacologia
11.
Med Res Rev ; 40(3): 1117-1141, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31793026

RESUMO

Since being discovered in 2008, the STING (stimulator of interferon genes) pathway has gradually been recognized as a central and promising target for immunotherapy. The STING pathway can be stimulated by cyclic dinucleotides (CDNs), leading to the type I interferons (IFN) production for immunotherapy for cancer or other diseases. However, the negative charges, hydrophilicity, and instability of CDNs have hindered their further applications. In addition, chronic activation of the STING pathway has been found to be involved in autoimmune diseases as IFN overproduction. Thus, research and development of STING agonists and inhibitors has been a hot field for the treatment of several diseases. The past several years, especially 2018, has seen increasingly rapid advances in this field. Here, this review summarizes the synthesis and modification of CDNs, the identification of nonnucleotide agonists, the recent progress in delivery systems and the medical applications, such as personalized vaccine adjuvants, in detail. In addition, in this review, we summarize the STING inhibitors' advances from two aspects, covalent, and noncovalent inhibitors.


Assuntos
Imunoterapia/métodos , Proteínas de Membrana/agonistas , Proteínas de Membrana/antagonistas & inibidores , Animais , Cátions , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Citosol/metabolismo , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Humanos , Hidrogéis/química , Concentração de Íons de Hidrogênio , Lipossomos/química , Camundongos , Neoplasias/imunologia , Neoplasias/terapia , Oligonucleotídeos Fosforotioatos/química , Polímeros/química
12.
ACS Omega ; 4(5): 8874-8880, 2019 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-31459975

RESUMO

A series of thiophene derivatives were synthesized by functionalization of 2,3-fused thiophene scaffolds. Their cytotoxicity was assessed against HeLa and Hep G2 cells. Compound 480 was identified as a promising candidate because of its low IC50 in HeLa (12.61 µg/mL) and Hep G2 (33.42 µg/mL) cells. The drug was loaded into folic acid (FA)-coated nanoparticles (NPs) to address its poor water solubility and to improve its selectivity for cancer cells. Compound 480 was shown to induce apoptosis by changes in mitochondrial membrane potential (ΔΨm) and the reactive oxygen species level. Furthermore, FA-modified NPs enhanced uptake capacity compared to unmodified controls by flow cytometry. This drug delivered in folate nanocarriers is promising for the treatment of cancers.

13.
Molecules ; 24(1)2019 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-30621360

RESUMO

A series of thiophene derivatives (TPs) were synthesized and evaluated for cytotoxicity in HepG2 and SMMC-7721 cell lines by MTT assay. TP 5 was identified as a potential anticancer agent based on its ability to inhibit tumor cell growth. Drawbacks of TPs, including poor solubility and high toxicity, were overcome through delivery using self-assembling HSA nanoparticles (NPs). The optimum conditions for TP 5-NPs synthesis obtained by adjusting the temperature and concentration of TP 5. The NPs had an encapsulation efficiency of 99.59% and drug-loading capacity of 3.70%. TP 5 was slowly released from TP 5-NPs in vitro over 120 h. HepG2 and SMMC-7721 cell lines were employed to study cytotoxicity of TP 5-NPs, which exhibited high potency. ROS levels were elevated and mitochondrial membrane potentials reversed when the two cell lines were treated with TP 5-NPs for 12 h. Cellular uptake of fluorescence-labeled TP 5-NPs in vitro was analyzed by flow cytometry and laser confocal scanning microscopy. Fluorescence intensity increased over time, suggesting that TP 5-NPs were efficiently taken up by tumor cells. In conclusion, TP 5-NPs showed great promise as an anticancer therapeutic agent.


Assuntos
Albuminas/química , Antineoplásicos/farmacologia , Portadores de Fármacos/química , Nanopartículas/química , Tiofenos/farmacologia , Transporte Biológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada/química , Desenho de Fármacos , Liberação Controlada de Fármacos , Humanos , Estrutura Molecular , Tamanho da Partícula , Espécies Reativas de Oxigênio/metabolismo , Solubilidade , Relação Estrutura-Atividade , Propriedades de Superfície
14.
Acta Biochim Pol ; 65(3): 465-470, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30188965

RESUMO

Inflammation plays an important role in the development of many obesity-related diseases. This study aimed to investigate the effect of ezetimibe on inflammation and myocardial remodeling in obese rats. A rat model of obesity was established, and myocardial damage was examined by transmission electron microscopy and Masson staining. Twenty obese rats were divided into two groups (n=10): obese group and ezetimibe group. Ten SD rats were used as controls. Western blot was performed to monitor the expression of P-p38MAPK and interleukin (IL)-6. Immunohistochemical staining was used to monitor the expression of intercellular adhesion molecule-1 and vascular cell adhesion molecule-1. In the obese rats group, we observed increased inflammatory factors and myocardial hypertrophy. In contrast, the ezetimibe group exhibited decreased expression of inflammatory factors and an improvement in myocardial remodeling compared to the obese group. Mechanistically, we found that ezetimibe decreased P-p38MAPK, IL-6, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 levels in the hearts of the obese rats. Taken together, these results indicate that ezetimibe may improve myocardial remodeling in obese rats by inhibiting inflammation.


Assuntos
Anticolesterolemiantes/farmacologia , Ezetimiba/farmacologia , Coração/efeitos dos fármacos , Inflamação/prevenção & controle , Obesidade/fisiopatologia , Remodelação Ventricular/efeitos dos fármacos , Animais , Western Blotting , Proteína C-Reativa/metabolismo , Cardiomegalia/etiologia , Colágeno/metabolismo , Dieta Hiperlipídica , Inflamação/complicações , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/metabolismo , Masculino , Miocárdio/metabolismo , Obesidade/complicações , Ratos Sprague-Dawley , Molécula 1 de Adesão de Célula Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Cell Physiol Biochem ; 48(5): 1843-1853, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30092566

RESUMO

BACKGROUND/AIMS: Consumption of a high-fat (HF) diet exacerbates metabolic cardiomyopathy through lipotoxic mechanisms. In this study, we explored the role of aldehyde dehydrogenase-2 (ALDH2) in myocardial damage induced by a HF diet. METHODS: Wild-type C57 BL/6J mice were fed a HF diet or control diet for 16 weeks. ALDH2 overexpression was achieved by injecting a lentiviral ALDH2 expression vector into the left ventricle. RESULTS: Consumption of a HF diet induced metabolic syndrome and myocardial remodeling, and these deleterious effects were attenuated by ALDH2 overexpression. In addition, ALDH2 overexpression attenuated the cellular apoptosis and insulin resistance associated with a HF diet. Mechanistically, ALDH2 overexpression inhibited the expression of c-Jun N-terminal kinase (JNK)-1, activated protein 1 (AP-1), insulin receptor substrate 1 (IRS-1), 4- hydroxynonenal, caspase 3, transforming growth factor ß1, and collagen I and III, and enhanced Akt phosphorylation. CONCLUSION: ALDH2 may effectively attenuate myocardial remodeling and contractile defects induced by a HF diet through the regulation of the JNK/AP-1 and IRS-1/Akt signaling pathways. Our study demonstrates that ALDH2 plays an essential role in protecting cardiac function from lipotoxic cardiomyopathy.


Assuntos
Aldeído-Desidrogenase Mitocondrial/metabolismo , Dieta Hiperlipídica , Miocárdio/metabolismo , Aldeído-Desidrogenase Mitocondrial/genética , Animais , Apoptose , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Potencial da Membrana Mitocondrial , Síndrome Metabólica/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Contração Miocárdica , Miocárdio/patologia , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Remodelação Ventricular
16.
Chem Commun (Camb) ; 54(69): 9655-9658, 2018 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-30101273
17.
Biosens Bioelectron ; 99: 28-33, 2018 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-28735043

RESUMO

In this work, we have developed an electrochemical aptasensor for high-sensitivity determination of carcinoembryonic antigen (CEA) based on lead ion (Pb2+)-dependent DNAzyme-assisted signal amplification and graphene quantum dot-ionic liquid-nafion (GQDs-IL-NF) composite film. We designed hairpin DNA containing CEA-specific aptamers and DNAzyme chains. In the presence of CEA, hairpin DNA recognized the target and performed a DNAzyme-assisted signal amplification reaction to yield a large number of single-stranded DNA. The GQDs-IL-NF composite film was immobilized on the glassy carbon electrode for the interaction with single-stranded DNA through noncovalent π-π stacking interaction. Therefore, the methylene blue-labeled substrate DNA (MB-substrate) was fixed on the electrode and exhibited an initial electrochemical signal. Under optimal conditions, the response current change was proportional to the concentration of CEA, demonstrating a wide linear range from 0.5fgmL-1 to 0.5ngmL-1, with a low detection limit of 0.34fgmL-1. Furthermore, the proposed aptasensor was successfully applied in determining CEA in serum samples, showing its superior prospects in clinical diagnosis.


Assuntos
Aptâmeros de Nucleotídeos/química , Técnicas Biossensoriais , Antígeno Carcinoembrionário/isolamento & purificação , DNA Catalítico/química , Antígeno Carcinoembrionário/sangue , DNA de Cadeia Simples/química , Polímeros de Fluorcarboneto/química , Ouro/química , Grafite/química , Humanos , Líquidos Iônicos , Limite de Detecção , Nanoestruturas/química , Pontos Quânticos/química
20.
Inorg Chem ; 50(4): 1304-8, 2011 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-21254752

RESUMO

Reactions of lanthanide(III) perchlorate (Ln = Dy, Tb, and Gd), nickel(II) acetate, and ditopic ligand 2-(benzothiazol-2-ylhydrazonomethyl)-6-methoxyphenol (H(2)L) in a mixture of methanol and acetone in the presence of NaOH resulted in the successful assembly of novel Ln(2)Ni(2) heterometallic clusters representing a new heterometallic 3d-4f motif. Single-crystal X-ray diffraction reveals that all compounds are isostructural, with the central core composed of distorted [Ln(2)Ni(2)O(4)] cubanes of the general formula [Ln(2)Ni(2)(µ(3)-OH)(2)(OH)(OAc)(4)(HL)(2)(MeOH)(3)](ClO(4))·3MeOH [Ln = Dy (1), Tb (2), and Gd (3)]. The magnetic properties of all compounds have been investigated. Magnetic analysis on compound 3 indicates ferromagnetic Gd···Ni exchange interactions competing with antiferromagnetic Ni···Ni interactions. Compound 1 displays slow relaxation of magnetization, which is largely attributed to the presence of the anisotropic Dy(III) ions, and thus represents a new discrete [Dy(2)Ni(2)] heterometallic cubane exhibiting probable single-molecule magnetic behavior.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA