Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(27): eado4719, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968352

ABSTRACT

Solid-state electrolytes (SSEs) are challenged by complex interfacial chemistry and poor ion transport through the interfaces they form with battery electrodes. Here, we investigate a class of SSE composed of micrometer-sized lithium oxide (Li2O) particles dispersed in a polymerizable 1,3-dioxolane (DOL) liquid. Ring-opening polymerization (ROP) of the DOL by Lewis acid salts inside a battery cell produces polymer-inorganic hybrid electrolytes with gradient properties on both the particle and battery cell length scales. These electrolytes sustain stable charge-discharge behavior in Li||NCM811 and anode-free Cu||NCM811 electrochemical cells. On the particle length scale, Li2O retards ROP, facilitating efficient ion transport in a fluid-like region near the particle surface. On battery cell length scales, gravity-assisted settling creates physical and electrochemical gradients in the hybrid electrolytes. By means of electrochemical and spectroscopic analyses, we find that Li2O particles participate in a reversible redox reaction that increases the effective CE in anode-free cells to values approaching 100%, enhancing battery cycle life.

2.
Cancers (Basel) ; 15(21)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37958443

ABSTRACT

The aim of the present study was to evaluate the effect of ETS homologous factor (EHF) in malignant breast cancer cells. The overexpression and knockdown of the EHF gene in human and mouse breast cancer cells were performed, and the TCGA dataset and Q-omics were analyzed. We found that the tumor suppressor NDRG2 is correlated with EHF gene expression in triple-negative breast cancer cells, that EHF overexpression results in reduced cell proliferation and that apoptosis is promoted by the chemotherapeutic reagent treatment of EHF-overexpressing cells. By EHF overexpression, senescence-associated ß-galactosidase activity and p21WAF1/CIP1 expression were increased, suggesting that EHF may induce cellular senescence. In addition, the overexpression of EHF reduced the migratory ability and inhibited epithelial-mesenchymal transition (EMT). Furthermore, EHF inhibited the phosphorylation of STAT3. The overexpression of EHF also reduced the tumor size, and lung metastasis in vivo. At the tumor site, ß-galactosidase activity was increased by EHF. Finally, the Kaplan-Meier-plotter analysis showed that TNBC patients with a high expression of EHF had a longer relapse-free survival rate. Our findings demonstrated that EHF inhibits breast tumor progression by inducing senescence and regulating EMT in TNBC cells.

3.
ChemSusChem ; 15(5): e202102394, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35132831

ABSTRACT

A full cell chemistry of aqueous dual-ion battery (DIB) was reported, comprising the graphite cathode and 3,4,9,10-perylenetetracarboxylic diimide (PTCDI) as the anode. This DIB employed a mixture aqueous electrolyte: 5 m tributylmethylammonium (TBMA) chloride plus 5 m MgCl2 , where [MgCl3 ]- and TBMA+ serve as the charge carriers for cathode and anode of the DIB, respectively. This novel full cell exhibited a specific capacity of around 41 mAh g-1 based on the total active mass of both electrodes with an average operation voltage of 1.45 V and stable cycling for 400 cycles.

4.
Angew Chem Int Ed Engl ; 59(45): 19924-19928, 2020 Nov 02.
Article in English | MEDLINE | ID: mdl-32710468

ABSTRACT

Oxidative anion insertion into graphite in an aqueous environment represents a significant challenge in the construction of aqueous dual-ion batteries. In dilute aqueous electrolytes, the oxygen evolution reaction (OER) dominates the anodic current before anions can be inserted into the graphite gallery. Herein, we report that the reversible insertion of Mg-Cl superhalides in graphite delivers a record-high reversible capacity of 150 mAh g-1 from an aqueous deep eutectic solvent comprising magnesium chloride and choline chloride. The insertion of Mg-Cl superhalides in graphite does not form staged graphite intercalation compounds; instead, the insertion of Mg-Cl superhalides makes the graphite partially turbostratic.

5.
Quant Imaging Med Surg ; 10(3): 789-797, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32269937

ABSTRACT

Streptozotocin treatment has emerged as an alternative model of sporadic Alzheimer's disease (SAD). Streptozotocin-induced alterations in iron and calcium levels reflect magnetic susceptibility changes, while susceptibility distribution in the cerebral regions has not been reported yet. This study aimed to investigate susceptibility distribution in the limbic system after streptozotocin administration to cynomolgus monkeys for exploring informative SAD biomarkers. Quantitative susceptibility mapping (QSM) using 7T magnetic resonance imaging (MRI) was utilized to quantitatively compare the susceptibility distributions in monkeys with sporadic Alzheimer disease and age-matched healthy controls. Compared to healthy controls, overall susceptibility values differed in the SAD models. Notable substantial susceptibility changes were observed in the hypothalamus with a 4.38-time decrease (AD: -47.45±12.19 ppb, healthy controls: 14.02±9.51 ppb) and in the posterior parts of the corpus callosum with a 2.83-times increase (AD: 31.49±15.90 ppb; healthy controls: 11.13±4.02 ppb). These susceptibility alterations may reflect neuronal death, and could serve as key biomarkers in the SAD. These results may be useful for specifying AD pathologies such as cognitive and non-cognitive symptoms.

6.
Oncotarget ; 9(24): 16744-16757, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29682182

ABSTRACT

Angelica amurensis has traditionally been used to treat various medical problems. In this report, we introduce cis-khellactone as a new anti-cancer agent, which was isolated from the chloroform soluble fraction of the rhizomes of Angelica amurensis. Its anti-cancerous effect was at first tested in MCF7 and MDA-MB-231 breast cell lines, in which MCF7 is well known to be resistant to many anti-cancer drugs; MCF10A normal breast cell line was used as a control. In vitro experiments showed that cis-khellactone suppressed cell growth and proliferation at a relatively low concentrations (<5 µg/ml) and decreased cell viability at high concentrations (>10 µg/ml) in both cancer cell lines in a time- and concentration-dependent manner. This anti-cancerous effect was also checked in additional 16 different types of normal and cancer cell lines. Cis-khellactone treatment significantly suppressed cell proliferation and enhanced cell death in all tested cancer cell lines. Furthermore, Western blot analysis showed that cis-khellactone induced three types of programmed cell death (PCD): apoptosis, autophagy-mediated cell death, and necrosis/necroptosis. Cis-khellactone concentration-dependently decreased cell viability by increasing the level of reactive oxygen species (ROS) and decreasing mitochondrial membrane potential (MMP), which are related to all three types of PCD. Mitochondrial fractionation data revealed that cis-khellactone induced the translocation of BAX and BAK into mitochondria as well as the overexpression of VDAC1, which probably accelerates MMP disruption and finally cell death. Importantly, our extended in vivo studies with xenograft model further confirmed these findings of anti-cancerous effects and showed no harmful effects in normal tissues, suggesting that there would be no side effects in humans.

7.
J Control Release ; 261: 307-317, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28642155

ABSTRACT

Small, polar compounds, both ionic and uncharged, partition into human stratum corneum immersed in aqueous solutions to an extent comparable to the water volume fraction of the tissue, then desorb in two phases. The fast phase has a time constant on the order of a few minutes, whereas the slow phase occurs over many hours. A physical model for this behavior involving a combination of tranverse diffusion through the tissue and lateral diffusion and exchange with skin appendages is presented. This concept is probed using excised human stratum corneum exposed to aqueous solutions of radiolabeled sodium chloride, tetraethyl ammonium bromide and mannitol, plus previously published data on six other compounds of varying molecular size and polarity. The fast phase desorption process becomes unimportant for lipophilic compounds. Slow phase desorption rates are size-selective, with larger species desorbing much more slowly than smaller ones. Interpreting the size-selectivity in terms of smooth cylindrical pores using the centerline approximation leads to an optimum pore radius of about 8-12Å, depending on the model chosen.


Subject(s)
Mannitol/pharmacokinetics , Skin Absorption , Sodium Chloride/pharmacokinetics , Tetraethylammonium/pharmacokinetics , Biological Transport , Diffusion , Humans , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Mannitol/chemistry , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Skin/metabolism , Sodium Chloride/chemistry , Tetraethylammonium/chemistry , Water/chemistry
8.
Int J Oncol ; 48(6): 2639-46, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27035851

ABSTRACT

TRIP-Br1 oncoprotein is known to be involved in many vital cellular functions. In this study, we examined the role of TRIP-Br1 in hypoxia-induced cell death. Exposure to the overcrowded and CoCl2-induced hypoxic conditions increased TRIP-Br1 expression at the protein level in six breast cancer cell lines (MCF7, MDA-MB-231, T47D, Hs578D, BT549, and MDA-MB-435) but resulted in no significant change in three normal cell lines (MCF10A, MEF and NIH3T3). Our result revealed that CoCl2-induced hypoxia stimulated apoptosis and autophagy, in which TRIP-Br1 expression was found to be upregulated. Interestingly, TRIP-Br1 silencing in the MCF7 and MDA-MB-231 cancer cells accelerated apoptosis and destabilization of XIAP under the CoCl2-induced hypoxic condition, implying that TRIP-Br1 may render cancer cells resistant to apoptosis through the stabilization of XIAP. We also propose that TRIP-Br1 seems to be upregulated at least partly as a result of the inhibition of PI3K/AKT signaling pathway and the overexpression of HIF-1α. In conclusion, our findings suggest that TRIP-Br1 functions as an oncogenic protein by providing cancer cells resistance to the hypoxia-induced cell death.


Subject(s)
Breast Neoplasms/metabolism , Cobalt/toxicity , Down-Regulation , Drug Resistance, Neoplasm , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Apoptosis , Cell Hypoxia , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Transcription Factors , Up-Regulation , X-Linked Inhibitor of Apoptosis Protein/metabolism
9.
Oncotarget ; 6(30): 29060-75, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26334958

ABSTRACT

TRIP-Br1 oncogenic protein has been shown to have multiple biological functions in cells. In this study, we demonstrate that TRIP-Br1 functions as an oncoprotein by inhibiting autophagy, apoptosis, and necroptosis of cancer cells and eventually helping them to survive under the nutrient/serum starved condition. TRIP-Br1 expression level was significantly increased in conditions with low levels of nutrients. Nutrient depleted conditions were induced by culturing cancer cells until they were overcrowded with high cell density or in media deprived of glucose, amino acids, or serum. Among them, serum starvation significantly enhanced the expression of TRIP-Br1 only in all tested breast cancer cell lines (MCF7, MDA-MB-231, T47D, MDA-MB-435, Hs578D, BT549, and MDA-MB-435) but not in the three normal cell lines (MCF10A, HfCH8, and NIH3T3). As compared with the control cells, the introduction of TRIP-Br1 silencing siRNA into MCF7 and MDA-MB-231 cells accelerated cell death by inducing apoptosis and necroptosis. In this process, TRIP-Br1 confers resistance to serum starvation-induced cell deaths by stabilizing the XIAP protein and inhibiting cellular ROS production. Moreover, our data also show that the intracellular increase of TRIP-Br1 protein resulting from serum starvation seems to occur in part through the blockage of PI3K/AKT signaling pathway.


Subject(s)
Amino Acids/deficiency , Apoptosis , Autophagy , Breast Neoplasms/metabolism , Glucose/deficiency , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Survival , Culture Media, Serum-Free/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Necrosis , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction , Time Factors , Trans-Activators/genetics , Transcription Factors , Transfection , X-Linked Inhibitor of Apoptosis Protein/metabolism
10.
Oncotarget ; 6(10): 7522-35, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25691055

ABSTRACT

TRIP-Br3 and TRIP-Br1 have shown to have important biological functions. However, the function of TRIP-Br3 in tumorigenesis is not well characterized compared to oncogenic TRIP-Br1. Here, we investigated the function of TRIP-Br3 in tumorigenesis by comparing with that of TRIP-Br1. Under nutrient/serum starvation, TRIP-Br3 expression was down-regulated slightly in cancer cells and significantly in normal cells. Unexpectedly, TRIP-Br1 expression was greatly up-regulated in cancer cells but not in normal cells. Moreover, TRIP-Br3 activated autophagy while TRIP-Br1 inactivated it under serum starvation. In spite of different expression and roles of TRIP-Br3 and TRIP-Br1, both of them alleviate cell death by directly binding to and stabilizing XIAP, a potent apoptosis inhibitor, through blocking its ubiquitination. Taken together, we propose that TRIP-Br3 primarily activates the autophagy and suppresses apoptosis in nutrient sufficient condition. However, the prolonged extreme stressful condition of nutrient starvation causes a dramatic decrease of TRIP-Br3, which in turn induces apoptosis by destabilizing XIAP. Up-regulated TRIP-Br1 in cancer cells compensates this effect and delays apoptosis. This can be explained by the competitive alternative binding of TRIP-Br3 and TRIP-Br1 to the BIR2 domain of XIAP. In an extended study, our immunohistochemical analysis revealed a markedly lower level of TRIP-Br3 protein in human carcinoma tissues compared to normal epithelial tissues, implying the role of TRIP-Br3 as a tumor suppressor rather than onco-protein.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Apoptosis , Down-Regulation , Humans , Transfection , X-Linked Inhibitor of Apoptosis Protein/analysis
11.
Int J Oncol ; 45(1): 189-96, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24789658

ABSTRACT

The p34(SEI-1) oncoprotein is involved in a transcriptional regulation, cell cycle regulation, apoptosis, development and many other important cellular functions. Our present study suggests that p34(SEI-1) can promote metastasis by enhancing migration and invasion of cancer cells. Consistently, p34(SEI-1) expression was found to be increased as the tumor invasiveness progressed in human breast tissues. p34(SEI-1) may promote cancer metastasis by activating the PI3K/AKT signaling pathway. In this process, p34(SEI-1) activates two different serine/threonine kinases, AKT or ILK, depending on the expression status of HER2/neu oncogene. In HER2/neu suppressed cancer cells, p34(SEI-1) promoted metastasis mainly by activating AKT via phosphorylation of the 473 serine residue. In HER2/neu expressing cancer cells, p34(SEI-1) overexpression downregulates HER2/neu expression, leading to the activation of another crucial serine/threonine kinase ILK due to phosphorylation of the 178 threonine residue instead of AKT. These results suggest that p34(SEI-1) affects cancer metastasis by regulating two different signaling pathways depending on the HER2/neu expression level, in which AKT and ILK modulation can be stimulated by p34(SEI-1) overexpression.


Subject(s)
Breast Neoplasms/pathology , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Trans-Activators/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Female , HEK293 Cells , Humans , MAP Kinase Signaling System , MCF-7 Cells , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Trans-Activators/genetics , Transcription Factors
12.
Int J Oncol ; 43(5): 1587-95, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23970032

ABSTRACT

A 34-KD protein encoded by the SEI-1 gene (p34(SEI­1)), is a relatively recently discovered oncoprotein that has multiple important biological functions. Our data show that p34(SEI-1) enhances cancer cell survival and promotes tumorigenesis by downregulating the tumor suppressor PTEN, a negative regulator of the PI3K/AKT signaling pathway, and therefore activating the PI3K/AKT signaling pathway. In this process, p34(SEI-1) positively affects NEDD4-1 gene expression both at the transcriptional and protein levels. Furthermore, the expression levels of p34(SEI-1) and NEDD4-1 were found to be coordinated in tumor tissues obtained from patients with breast cancer. We also show that p34(SEI-1) affects the subcellular localization of PTEN.


Subject(s)
Breast Neoplasms/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/metabolism , Ubiquitin-Protein Ligases/metabolism , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/genetics , Female , Humans , Immunoenzyme Techniques , Nedd4 Ubiquitin Protein Ligases , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Proteolysis , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transcription Factors , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination
13.
Int J Oncol ; 32(2): 435-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202766

ABSTRACT

Recent studies have suggested that Skp2, an SCF-type ubiquitin ligase, positively regulates cell cycle through degradation of p27, which is an inhibitor of cyclin-dependent kinase 2 (CDK2), which drives cells from the G1 to S phase of cell cycles. In the present study, we examined key regulatory proteins involved in serum starvation-induced cell cycle arrest in human ovarian cancer cells, SK-OV-3. Cell cycle analysis showed that cells were arrested at the G1 phase after serum starvation. Western blot analysis showed that the protein levels of CDK4 and CDK2 were significantly decreased in SK-OV-3 cells. Consistently, Roscovitine, an inhibitor of CDK2, induced cell cycle arrest in normally proliferating cells and a chemical inhibitor of CDK4, 3-ATA [3-Amino-9-thio(10H)-acridone], was found to induce growth arrest. We also found that the protein level of Skp2 was dramatically decreased in response to serum starvation. Moreover, CDK2 protein, which allows cell cycle transit from the G1 to the S phase, was decreased when the Skp2 expression was inhibited by specific siRNA of Skp2, but CDK4 was not decreased. Therefore, these results suggest that serum starvation induces G1 arrest through suppression of Skp2-dependent CDK2 activity and Skp2-independent CDK4 activity in human SK-OV-3 ovarian cancer cells.


Subject(s)
Culture Media, Serum-Free/pharmacology , Cyclin-Dependent Kinase 2/biosynthesis , Cyclin-Dependent Kinase 4/biosynthesis , G1 Phase , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/metabolism , S-Phase Kinase-Associated Proteins/biosynthesis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Humans , Ovarian Neoplasms/drug therapy , RNA Interference , RNA, Small Interfering/metabolism , Time Factors
14.
Oncol Rep ; 18(4): 811-5, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17786340

ABSTRACT

Although ascorbate (Vitamin C) has been shown to inhibit cell growth and induce cell death in variety of cancer cells, results reported in other studies are inconsistent with this conclusion. It was previously reported that ascorbate induces apoptosis in human breast cancer cells. However, the molecular mechanism for this is not clear. In this study, we demonstrate that ascorbate induces cell death through the apoptosis-inducing factor (AIF) in the human breast cancer cell lines, SK-BR3 and Hs578T, but not in a normal breast cell line, Hs578. Ascorbate treatment caused the nuclear translocation of AIF, which is retained in the mitochondria in healthy cells, but caspase cleavage is not induced. Moreover, MG132, an inhibitor of AIF release from mitochondria, blocked the induction of cell death. Furthermore, cells that had been treated with human AIF-specific siRNA resisted cell death induced by ascorbate, implying that the translocation of AIF from mitochondria to the nucleus is responsible for ascorbate-mediated cell death. Therefore, these results suggest that ascorbate activates a caspase-independent and AIF-mediated cell death pathway in human breast cancer cells, SK-BR3, and Hs578T.


Subject(s)
Antioxidants/pharmacology , Apoptosis Inducing Factor/metabolism , Apoptosis/drug effects , Ascorbic Acid/pharmacology , Breast Neoplasms/pathology , Antineoplastic Agents/pharmacology , Apoptosis Inducing Factor/antagonists & inhibitors , Apoptosis Inducing Factor/genetics , Breast Neoplasms/metabolism , Caspase Inhibitors , Caspases/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Fluorescent Antibody Technique , Humans , Immunoblotting , Leupeptins/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Protein Transport , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...