Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biosens Bioelectron ; 175: 112848, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33257184

RESUMO

Herein, we present a target-triggered bidirectional one-dimensional (1D) DNA walking nanomachine, built from a well-designed track, which could simultaneously move two different DNA walkers to the opposite direction along the track and release payload. This track is composed of a DNA walker station (chain S3) in the middle of track for storing two kinds of DNA walker (W1 and W2), and corresponding two kinds of payload conjugated DNA stators (chain S1, S2 and S4, S5) for the moving of walker on the two flanks of chain S3 respectively. Moreover, the chain S3 also serves as a target-assisted amplification platform based on a catalytic hairpin assembly (CHA)-like strategy. In the presence of target (nucleic acid), the dynamic assembly between hairpin (HP) and S3 is triggered for multiple recycling of target and releasing of W1 and W2. Since the W1 and W2 respectively correspond to 8-17 DNAzyme and 10-23 DNAzyme, they could cleave the RNA substrates with sequence specificity to move towards two opposite directions along the track at the same time, accompanying the release of payloads. Such a 1D DNA walking nanomachine is not only could propel the walker to move in two directions respectively but also improve the locomotion efficiency compared to the traditional single-directional 1D DNA walking nanomachine with the same amounts of stators. This concept of inducing the locomotion manner change on a 1D DNA device may provide a thought to facilitate the development of DNA dynamic nanomachines and intelligent nanosensors.


Assuntos
Técnicas Biossensoriais , DNA Catalítico , Nanopartículas Metálicas , DNA , Ouro
2.
Int Immunopharmacol ; 89(Pt A): 106999, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33045563

RESUMO

Cisplatin is widely used as a chemotherapeutic agent for treating patients with solid tumors. The most common side effect of cisplatin treatment is nephrotoxicity. Recent studies have shown that mitochondrial apoptotic pathways are involved in cisplatin-induced acute kidney injury (Cis-AKI). LIGHT, the 14th member of the tumor necrosis factor superfamily (TNFSF14), was found to induce apoptosis of certain types of tumor cells. So far, a link between LIGHT and Cis-AKI has not been reported. In this study, we observed that expression of LIGHT and its receptors HVEM and LTßR was increased in kidney tissues of mice after cisplatin treatment. LIGHT deficiency aggravated kidney injury, as evidenced by more severe tubular injury; remarkably increased levels of serum creatinine (Scr), blood urea nitrogen (BUN), and both kidney injury molecule-1 (KIM-1) and inflammatory cytokine mRNAs in renal tissues. Moreover, in the renal tissues of LIGHT KO mice, cisplatin-induced mitochondrion injury and the levels of the pro-apoptotic molecules Bax, Cytochrome C (Cyt C), cleaved caspase-3, and cleaved caspase-9 were dramatically increased; in contrast, the expression of anti-apoptotic molecule Bcl-2 was markedly reduced, compared to those in WT mice, suggesting that LIGHT deficiency accelerated cisplatin-induced mitochondrial apoptosis of renal tubular cells in these mice. Accordingly, treatment with recombinant human LIGHT (rLIGHT) was shown to alleviate cisplatin-induced kidney injury in vivo. Similar results were observed after the human renal tubular epithelial cell line HK-2 cells exposure to rLIGHT stimulation, evidenced by the reduction in the mitochondrion dysfunction (as confirmed by the significant reduced oxidative stress and membrane potential changes) and in the percentage of cells apoptosis. While blocking LIGHT with the soluble fusion protein LTßR-Ig or HVEM-Ig accelerated the HK-2 cells apoptosis. In conclusion, LIGHT deficiency aggravates Cis-AKI by promoting mitochondrial apoptosis pathways.


Assuntos
Injúria Renal Aguda/metabolismo , Apoptose , Cisplatino , Túbulos Renais/metabolismo , Mitocôndrias/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Túbulos Renais/patologia , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
3.
J Cell Mol Med ; 24(20): 11936-11948, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32881263

RESUMO

Sepsis-associated acute kidney injury (SA-AKI) is a common clinical critical care syndrome. It has received increasing attention due to its high morbidity and mortality; however, its pathophysiological mechanisms remain elusive. LIGHT, the 14th member of the tumour necrosis factor (TNF) superfamily and a bidirectional immunoregulatory molecule that regulates inflammation, plays a pivotal role in disease pathogenesis. In this study, mice with an intraperitoneal injection of LPS and HK-2 cells challenged with LPS were employed as a model of SA-AKI in vivo and in vitro, respectively. LIGHT deficiency notably attenuated kidney injury in pathological damage and renal function and markedly mitigated the inflammatory reaction by decreasing inflammatory mediator production and inflammatory cell infiltration in vivo. The TLR4-Myd88-NF-κB signalling pathway in the kidney of LIGHT knockout mice was dramatically down-regulated compared to the controls. Recombinant human LIGHT aggravated LPS-treated HK-2 cell injury by up-regulating the expression of the TLR4-Myd88-NF-κB signalling pathway and inflammation levels. TAK 242 (a selective TLR4 inhibitor) reduced this trend to some extent. In addition, blocking LIGHT with soluble receptor fusion proteins HVEM-Fc or LTßR-Fc in mice attenuated renal dysfunction and pathological damage in SA-AKI. Our findings indicate that LIGHT aggravates inflammation and promotes kidney damage in LPS-induced SA-AKI via the TLR4-Myd88-NF-κB signalling pathway, which provide potential strategies for the treatment of SA-AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Sepse/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Injúria Renal Aguda/patologia , Animais , Linhagem Celular , Regulação para Baixo , Humanos , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Camundongos Knockout , Modelos Biológicos , Análise de Sobrevida , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/deficiência
4.
Chem Sci ; 11(8): 2193-2199, 2020 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-34123311

RESUMO

Herein, we report a three-dimensional (3D) DNA walking nanomachine innovatively constructed from a functionalized 3D DNA track, which runs in an orderly manner with favorable directionality to allow for programming certain pathways of information transduction for some target tasks. The nanomachine was constructed using a departure station of walker (UB + W) and a functionalized 3D track, which was made up of a rolling circle amplification (RCA)-generated backbone chain and numerous triangular rung units with stators (UA + S) assembled into a repeating array along the backbone. A specific domain (SD) was designed at the 5'-end of the backbone to capture the UB + W, and stators with specific RNA substrates were immobilized at the three UA corners for the DNA walker to travel on. Powered by 10-23 DNAzyme, the DNA walker started moving from the SD end to the other end of the track by the autonomous cleavage of RNA substrates. Significantly, the homogeneous distribution of stators in the longitudinal and horizontal extensions paved a specific path for each walker to move along the 3D track. This resulted in random and inactive self-avoiding walking; thus, the nanomachine exhibited good executive ability. These properties allowed the DNA walking nanomachine to program the certain pathways of information transduction for the stepwise and programmed execution of some target tasks, such as the synthesis of specific polyorganics and cargo delivery. We believe that such a 3D DNA walking nanomachine could enrich the concept in the field of dynamic DNA nanotechnology, and may improve the development of novel DNA nanomachines in cargo delivery and composite product synthesis.

5.
Chin Med J (Engl) ; 119(13): 1088-93, 2006 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-16834927

RESUMO

BACKGROUND: Advanced oxidation protein products (AOPPs) are new uremic toxins reported by Witko-Sarsat in 1996, which are associated with the pathogenesis of atherosclerosis. However, the mechanisms by which AOPPs enhance atherosclerosis have not been fully understood. Monocyte chemoattractant protein-1 (MCP-1) is a chemokine which stimulates migration of monocytes and plays a critical role in the development of atherosclerosis. In this study, we investigated the effect of AOPPs on MCP-1 expression in cultured vascular smooth muscle cells (VSMCs). METHODS: VSMCs were cultured and then co-incubated with AOPP (200 micromol/L, 400 micromol/L) for different times with or without pretreatment with specific p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580. RT-PCR and Western blott were used to detect MCP-1 mRNA and protein expression at different time points after AOPP stimulation in rat smooth muscle cells. Western blot was used to detect the expression of phosphorylated p38 MAPK. RESULTS: Treatment of VSMC with AOPPs resulted in a significant increase of the expression of MCP-1 mRNA and protein in time- and dose-dependent manner, and could activated p38 MAPK. Pretreatment of VSMCs with SB203580 resulted in a dose-dependent inhibition of AOPPs-induced MCP-1 mRNA and protein expression. CONCLUSIONS: AOPPs can stimulate MCP-1 expression via p38 MAPK in VSMCs. This suggests that AOPPs might contribute to the formation of atherosclerosis through this proinflammatory effect.


Assuntos
Aterosclerose/etiologia , Quimiocina CCL2/genética , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas/metabolismo , Uremia/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Doenças Cardiovasculares/etiologia , Células Cultivadas , Ativação Enzimática , Imidazóis/farmacologia , Falência Renal Crônica/complicações , Masculino , Músculo Liso Vascular/citologia , Oxirredução , Piridinas/farmacologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...