Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Appl Mater Interfaces ; 15(8): 11131-11140, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36799618

ABSTRACT

Metal-organic frameworks (MOFs) have been extensively studied in host-guest chemistry by means of ultrahigh porosities, tunable channels, and component diversities. As the host matrix, MOFs exhibit immense potential in the preparation of single-phase white light-emitting (SPWLE) materials. Nonetheless, it is a great challenge that the size of the introduced guest molecules is limited by MOF pores, which affects the WLE optimization. In this work, two-dimensional (2D) MOFs are first utilized as the host matrices to simultaneously encapsulate red-green-blue fluorescent dyes for SPWLE. Various dyes@2D MOF composites with high-quality WLE performances and ultrathin nanosheet morphologies are directly assembled from 2D MOF precursors and dyes in high yields. Owing to the flexible interlamellar space of 2D MOFs, different types and sizes of guests can be easily introduced, which greatly expands the range of available MOF hosts and guests, making the WLE much more tunable. The strategy of employing 2D MOFs as the host matrices to introduce multicomponent dyes for SPWLE nanosheets resolves the restriction of MOF pores on the guest molecule size and opens a new avenue to rationally design and prepare SPWLE nanosheets that are highly solution-processable.

2.
Oncotarget ; 7(35): 56798-56810, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27462864

ABSTRACT

The abnormality of DNA methylation is one of the major epigenetic alterations in the human hepatocellular carcinoma (HCC). We have assessed the global genomic DNA methylation profiles in human HCC patients by using the Infinium Human Methylation27 BeadChip. A CpG loci of S100A8 was found to be significantly hypomethylated in HCC.Pooled meta-analysis of five validation public datasets demonstrated its methylation level was significantly lower for HCC compared to paired adjacent normal tissues. Quantitative pyrosequencing analysis also showed that the S100A8 methylation level was decreased in cancer tissues (31.90%±13.31%) than that in the paired adjacent normal tissues (65.33%±3.64%, p<0.01). The area under the ROC curve (AUC) value was 0.950 (p<0.01). Kaplan-Meier survival curves revealed that hypomethylation of S100A8 was associated with shortened overall survival (OS) and progression-free survival (PFS) (log rank p<0.05). Multivariate Cox proportional hazards model also indicated significantly shorter OS (HR, 1.709; 95 % CI, 1.127-2.591) and PFS (HR, 1.767; 95 % CI, 1.168-2.974) were observed in the low-methylation-level group compared to the high-methylation-level group. Furthermore, S100A8 overexpression in Huh7 and MHCC-97H hepatoma cell lines led to increased cell proliferation, migration, invasion, and tumor growth. These findings suggested S100A8 methylation to be served as potential diagnosis and prognosis marker for HCC. S100A8 also may play as a tumor promoter in HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Calgranulin A/chemistry , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Animals , Area Under Curve , Carcinoma, Hepatocellular/diagnosis , Cell Line, Tumor , CpG Islands , DNA Methylation , Disease-Free Survival , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Liver Neoplasms/diagnosis , Male , Mice , Mice, Inbred BALB C , Multivariate Analysis , Prognosis , Promoter Regions, Genetic , Proportional Hazards Models , Sequence Analysis, DNA , Treatment Outcome
3.
PLoS One ; 8(5): e62652, 2013.
Article in English | MEDLINE | ID: mdl-23671619

ABSTRACT

Although pterostilbene (PTE) has been shown to have potent antitumor activities against various cancer types, the molecular mechanisms of these activities remain unclear. In this study, we investigated the antitumor activity of PTE against human lung adenocarcinoma in vitro and in vivo and explored the role of the Notch1 signaling pathway in this process. PTE treatment resulted in a dose- and time-dependent decrease in the viability of A549 cells. Additionally, PTE exhibited strong antitumor activity, as evidenced not only by a reduced mitochondrial membrane potential (MMP) and a decreased intracellular glutathione content but also by increases in the apoptotic index and the level of reactive oxygen species (ROS). Furthermore, PTE treatment induced the activation of the Notch1 Intracellular Domain (NICD) protein and activated Hes1. DAPT (a gamma secretase inhibitor) and Notch1 siRNA prevented the induction of NICD and Hes1 activation by PTE treatment and sensitized the cells to PTE treatment. The down-regulation of Notch signaling also prevented the activation of pro-survival pathways (most notably the PI3K/Akt pathway) after PTE treatment. In summary, lung adenocarcinoma cells may enhance Notch1 activation as a protective mechanism in response to PTE treatment. Combining a gamma secretase inhibitor with PTE treatment may represent a novel approach for treating lung adenocarcinoma by inhibiting the survival pathways of cancer cells.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy , Receptor, Notch1/metabolism , Stilbenes/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Apoptosis , Cell Line, Tumor , Cell Shape , Cell Survival/drug effects , Dipeptides/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Glutathione/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
PLoS One ; 8(3): e57941, 2013.
Article in English | MEDLINE | ID: mdl-23483946

ABSTRACT

Previous studies have shown that the JAK2/STAT3 signaling pathway plays a regulatory role in cellular oxidative stress injury (OSI). In this study, we explored the role of the JAK2/STAT3 signaling pathway in hydrogen peroxide (H2O2)-induced OSI and the protective effect of melatonin against (H2O2)-induced injury in human umbilical vein endothelial cells (HUVECs). AG490 (a specific inhibitor of the JAK2/STAT3 signaling pathway) and JAK2 siRNA were used to manipulate JAK2/STAT3 activity, and the results showed that AG490 and JAK2 siRNA inhibited OSI and the levels of p-JAK2 and p-STAT3. HUVECs were then subjected to H2O2 in the absence or presence of melatonin, the main secretory product of the pineal gland. Melatonin conferred a protective effect against H2O2, which was evidenced by improvements in cell viability, adhesive ability and migratory ability, decreases in the apoptotic index and reactive oxygen species (ROS) production and several biochemical parameters in HUVECs. Immunofluorescence and Western blotting showed that H2O2 treatment increased the levels of p-JAK2, p-STAT3, Cytochrome c, Bax and Caspase3 and decreased the levels of Bcl2, whereas melatonin treatment partially reversed these effects. We, for the first time, demonstrate that the inhibition of the JAK2/STAT3 signaling pathway results in a protective effect against endothelial OSI. The protective effects of melatonin against OSI, at least partially, depend upon JAK2/STAT3 inhibition.


Subject(s)
Human Umbilical Vein Endothelial Cells/pathology , Janus Kinase 2/metabolism , Melatonin/pharmacology , Oxidative Stress/drug effects , Protective Agents/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Apoptosis/drug effects , Caspase 3/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Cytochromes c/metabolism , Glutathione Peroxidase/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Hydrogen Peroxide/toxicity , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/metabolism , Superoxide Dismutase/metabolism , Tyrphostins/pharmacology , bcl-2-Associated X Protein/metabolism
5.
Cell Signal ; 25(3): 615-29, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23219912

ABSTRACT

Previous studies have demonstrated that Notch signaling pathway plays a regulatory role in cellular oxidative stress injury (OSI). In this study, our aim was to explore the role of the Notch signaling pathway in hydrogen peroxide (H(2)O(2))-induced OSI and the protective effect of curcumin during (H(2)O(2))-induced injury in human umbilical vein endothelial cells (HUVECs). DAPT, a specific inhibitor of the Notch signaling pathway, and Notch1 siRNA were used to study Notch activity. Further, HUVECs were exposed to H(2)O(2) in the absence or presence of curcumin. DAPT and Notch1 siRNA significantly inhibited OSI and the expression of Notch1 and Hes1. Curcumin conferred a protective effect on the HUVECs against H(2)O(2), which was evidenced by improved cell viability, adhesive ability and migratory ability and a decreased apoptotic index, decreased production of reactive oxygen species (ROS) and a reduction in several biochemical parameters. Immunofluorescence and Western blotting analyses demonstrated that H(2)O(2) treatment upregulated the expression of Notch1, Hes1, Caspase3, Bax and cytochrome c downregulated the expression of Bcl2, and treatment with curcumin reversed these effects. We demonstrated for the first time that the inhibition of Notch signaling pathway imparts a protective effect against endothelial OSI. The protective effects of curcumin against OSI are at least in part dependent on Notch1 inhibition.


Subject(s)
Curcumin/pharmacology , Oxidative Stress/drug effects , Protective Agents/pharmacology , Receptor, Notch1/antagonists & inhibitors , Signal Transduction/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Cytochromes c/metabolism , Dipeptides/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/toxicity , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Transcription Factor HES-1 , Up-Regulation , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...