Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Langmuir ; 38(14): 4378-4388, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35353515

ABSTRACT

Semiconducting two-dimensional (2D) materials have potential applications as ultrathin optoelectronic materials. Therefore, being able to precisely modulate the band gap is useful to improving their applicability. Electron doping of the semiconducting materials is one of the successful techniques used to modulate their band gap. Silver nanoclusters (AgNCs) or gold nanoclusters (AuNCs) a few nanometers in size can generate a high density of highly energetic hot electrons with relatively long lifetimes when photoexcited. The optical band gap of 2D MoS2 nanosheets shows different responses when integrated with different amounts of AgNCs or AuNCs due to the electron doping effect. Introducing a small amount of the nanoclusters to the surface of a MoS2 nanosheet lowered its optical band gap. Further reduction of the optical band gap of MoS2 is obtained upon tripling the amount of integrated nanoclusters. Conversely, the optical band gap of MoS2 was increased when integrated with 5 times the concentration of AuNCs and AgNCs. The optical band gap of the MoS2 nanosheets was significantly increased when integrated with an even higher concentration of AuNCs or AgNCs. The magnitude of the shift of the optical band gap of MoS2 induced by AgNCs is higher than that induced by AuNCs because the energy of LUMO of the AgNCs is higher than that of the AuNCs.


Subject(s)
Gold , Metal Nanoparticles , Electrons , Molybdenum , Silver
2.
Life (Basel) ; 11(9)2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34575110

ABSTRACT

Despite the fact that accumulation of microglia, the resident macrophages of the central nervous system (CNS) are the main feature of glioblastoma, the role of microglia in the progression of glioma is still arguable. Based on the correlation of inflammation with tumor progression, in this study, we attempt to determine if peripheral inflammation aggravates glioma expansion and the activation of microglia associated with the tumor. Experimental animals were administered intraperitoneally by inflammagen lipopolysaccharide (LPS) for 7 days (LPS priming) before intracerebral implantation of glioma cells. Moreover, a reduced level of tumor necrosis factor receptor type 2 (TNFR2) that is restricted to immune cells, neurons, and microglia has been found in patients with glioblastoma through the clinic analysis of monocyte receptor expression. Thus, in addition to wildtype (WT) mice, heterogeneous TNFR2 gene deficiency (TNFR2+/-) mice and homogeneous TNFR2 gene knockout (TNFR2-/-) mice were used in this study. The results show that peripheral challenge by LPS, Iba1+- or CD11b+-microglia increase in numbers in the cortex and hippocampus of TNFR2-/- mice, when compared to WT or TNFR2+/- mice. We further conducted the intracerebral implantation of rodent glioma cells into the animals and found that the volumes of tumors formed by rat C6 glioma cells or mouse GL261 glioma cells were significantly larger in the cortex of TNFR2-/- mice when compared to that measured in LPS-primed WT or LPS-primed TNFR2+/- mice. Ki67+-cells were exclusively clustered in the tumor of LPS-primed TNFR2-/- mice. Microglia were also extensively accumulated in the tumor formed in LPS-primed TNFR2-/- mice. Accordingly, our findings demonstrate that aggravation of microglia activation by peripheral inflammatory challenge and a loss of TNFR2 function might lead to the promotion of glioma growth.

3.
Langmuir ; 37(36): 10720-10731, 2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34473512

ABSTRACT

The two characteristic absorption peaks of semiconducting two-dimensional tungsten disulfide (WS2) are red-shifted after integrating with gold nanocube (AuNC) arrays. The amount of the red shift is reduced when the AuNCs are coated with a high concentration of Pd. A negligible shift was observed in the absorption peaks of WS2 when smaller amounts of Pd are introduced to the surface of AuNCs. Conversely, the photoluminescence (PL) of WS2 is blue-shifted when measured on top of AuNCs and AuNCs coated with different amounts of Pd. AuNC-Pd Janus nanoparticles are prepared by depositing Pd atoms asymmetrically on AuNCs assembled into 2-D arrays on the surface of a glass substrate by the chemical reduction of Pd ions. Due to the large AuNC or AuNC-Pd/WS2 Schottky barrier, the plasmon-induced hot electron transfer (PHET) from AuNCs and AuNCs coated with a high concentration of Pd is responsible for the red shift of the absorption spectrum of WS2. Introducing a lower concentration of Pd to AuNCs increases the Schottky barrier further due to the formation of the Au-Pd equilibrium Fermi level of lower energy, reducing the efficiency of PHET. The effect of Pd on the Fermi level of AuNCs vanishes at high Pd deposition. Pauli blocking and phase-space filling are responsible for the blue shift of PL of WS2 on top of AuNCs and AuNCs coated with Pd. The Pauli blocking effect is directly proportional to the PHET efficiency. This explains the significant blue shift of PL of WS2 after integrating with AuNCs and AuNCs coated with a high concentration of Pd. Additionally, depositing Pd onto AuNCs elongates the lifetime of the hot electrons and enhances the PHET efficiency.


Subject(s)
Gold , Nanostructures , Disulfides , Palladium
4.
Antioxidants (Basel) ; 10(5)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068192

ABSTRACT

Early-life sleep deprivation (ESD) is a serious condition with severe cognitive sequelae. Considering hippocampus plays an essential role in cognitive regulation, the present study aims to determine whether melatonin, a neuroendocrine beard with significant anti-oxidative activity, would greatly depress the hippocampal oxidative stress, improves the molecular machinery, and consequently exerts the neuro-protective effects following ESD. Male weanling Wistar rats (postnatal day 21) were subjected to ESD for three weeks. During this period, the animals were administered normal saline or melatonin (10 mg/kg) via intraperitoneal injection between 09:00 and 09:30 daily. After three cycles of ESD, the animals were kept under normal sleep/wake cycle until they reached adulthood and were sacrificed. The results indicated that ESD causes long-term effects, such as impairment of ionic distribution, interruption of the expressions of neurotransmitters and receptors, decreases in the levels of several antioxidant enzymes, and impairment of several signaling pathways, which contribute to neuronal death in hippocampal regions. Melatonin administration during ESD prevented these effects. Quantitative evaluation of cells also revealed a higher number of neurons in the melatonin-treated animals when compared with the saline-treated animals. As the hippocampus is critical to cognitive activity, preserving or even improving the hippocampal molecular machinery by melatonin during ESD not only helps us to better understand the underlying mechanisms of ESD-induced neuronal dysfunction, but also the therapeutic use of melatonin to counteract ESD-induced neuronal deficiency.

5.
ACS Nano ; 15(6): 10241-10252, 2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34032116

ABSTRACT

Utilizing solar energy for chemical transformations has attracted a growing interest in promoting the clean and modular chemical synthesis approach and addressing the limitations of conventional thermocatalytic systems. Under light irradiation, noble metal nanoparticles, particularly those characterized by localized surface plasmon resonance, commonly known as plasmonic nanoparticles, generate a strong electromagnetic field, excited hot carriers, and photothermal heating. Plasmonic nanoparticles enabling efficient absorption of light in the visible range have moderate catalytic activities. However, the catalytic performance of a plasmonic nanoparticle can be significantly enhanced by incorporating a highly catalytically active metal domain onto its surface. In this study, we demonstrate that femtosecond laser-induced atomic redistribution of metal domains in bimetallic Au-Pd nanorods (NRs) can enhance its photocurrent response by 2-fold compared to parent Au-Pd NRs. We induce structure changes on Au-Pd NRs by irradiating them with a femtosecond pulsed laser at 808 nm to precisely redistribute Pd atoms on AuNR surfaces, resulting in modified electronic and optical properties and, thereby, enhanced catalytic activity. We also investigate the trade-off between the effect of light absorption and catalytic activity by optimizing the structure and composition of bimetallic Au-Pd nanoparticles. This work provides insight into the design of hybrid plasmonic-catalytic nanostructures with well-tailored geometry, composition, and structure for solar-fuel-based applications.

6.
Antioxidants (Basel) ; 9(11)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33202817

ABSTRACT

Long-term poor glycemic control negatively affects macrovascular and microvascular diseases, as well as wound restoration. Buckwheat is a good source of rutin (quercetin-3-O-rutoside) and has benefits in regulating blood sugar. This study was to evaluate the antioxidant and anti-inflammatory effects of rutin on wound healing in streptozotocin-induced hyperglycemic rats. Eighteen male Wistar rats were randomly divided into three groups: normal (NDM), hyperglycemic (DM), and hyperglycemic with rutin (DMR). After induction of hyperglycemia for 2 days, a 15 × 15 mm wound was induced on the back of each rat. Intraperitoneal injection of rutin significantly ameliorated diabetes-induced body weight loss and improved metabolic dysfunctions of hyperglycemic rats. Based on appearance and histopathological staining, rutin promotes wound healing and inhibits production of inflammatory cells. The immunoblotting data indicated that rutin promotes production of antioxidant enzymes induced by nuclear factor erythroid 2-related factor 2 (NRF2), inhibits the expression of matrix metalloproteinases (MMPs) regulated by NF-κB, and decreases the expression of vascular endothelial growth factor (VEGF). It also promotes the expression of neurogenic-related protein (UCH-L1). The aforementioned results indicated that rutin reduces oxidative stress and inflammatory response in hyperglycemic rats, promoting wound healing and subsequently reducing the risk of wound ulcers.

7.
Langmuir ; 36(10): 2574-2583, 2020 Mar 17.
Article in English | MEDLINE | ID: mdl-32090574

ABSTRACT

Ultra-high-resolution optical microscopic techniques are used to measure the reflectance and photoluminescence (PL) spectrum of individual monolayered MoS2 of dimensions below 200 × 200 nm, while placed on top of a thin film conjugated polymer (CP). p-type and n-type CPs such as poly(3-hexylthiophene-2,5-diyl) (P3HT) and [6,6]-phenyl C61 butyric acid methyl ester (PCBM), respectively, modified the optical band gap of the MoS2 sheet differently. However, the optical band gap is decreased after integration with P3HT, while it is increased after being combined with PCBM. The acceptable reason for the modification of the band gap of MoS2 by CPs is the generation of interlayer excitons (ILE) at their interface. The optical band gap of MoS2 is further changed by introducing an inert polymer spacer of different thickness to separate MoS2 from the CP. This is attributed to the reduction of the efficiency of excitonic interactions and lowering the exciton binding energy, which is induced by the increase of the dielectric function at the CP-MoS2 interface. No sign of electron injection to the conduction band of MoS2 after integration with P3HT or PCBM, as no significant shift of the A1' Raman band of MoS2 was measured on top of CPs, which is sensitive to the electron injection.

8.
Oncogenesis ; 9(2): 9, 2020 Feb 04.
Article in English | MEDLINE | ID: mdl-32019907

ABSTRACT

Chondroitin sulfate synthases, a family of enzyme involved in chondroitin sulfate (CS) polymerization, are dysregulated in various human malignancies, but their roles in glioma remain unclear. We performed database analysis and immunohistochemistry on human glioma tissue, to demonstrate that the expression of CHSY1 was frequently upregulated in glioma, and that it was associated with adverse clinicopathologic features, including high tumor grade and poor survival. Using a chondroitin sulfate-specific antibody, we showed that the expression of CHSY1 was significantly associated with CS formation in glioma tissue and cells. In addition, overexpression of CHSY1 in glioma cells enhanced cell viability and orthotopic tumor growth, whereas CHSY1 silencing suppressed malignant growth. Mechanistic investigations revealed that CHSY1 selectively regulates PDGFRA activation and PDGF-induced signaling in glioma cells by stabilizing PDGFRA protein levels. Inhibiting PDGFR activity with crenolanib decreased CHSY1-induced malignant characteristics of GL261 cells and prolonged survival in an orthotopic mouse model of glioma, which underlines the critical role of PDGFRA in mediating the effects of CHSY1. Taken together, these results provide information on CHSY1 expression and its role in glioma progression, and highlight novel insights into the significance of CHSY1 in PDGFRA signaling. Thus, our findings point to new molecular targets for glioma treatment.

9.
Langmuir ; 35(14): 4956-4965, 2019 Apr 09.
Article in English | MEDLINE | ID: mdl-30874438

ABSTRACT

Polyethylene glycol (PEG) assembled on the surface of two-dimensional tungsten disulfide (WS2) into a limited number of nanoislands (NIs), nanoshells (NSs), and granular nanoparticulates (GNPs) depending on its chain length. NI assemblies showed a nonmeasurable shift of photoluminescence (PL) and the A and B absorption peaks of WS2. This confirmed that the electronic doping by thiol is not effective. The PEG NS assembly displayed a smaller red shift of the PL and a slight decrease of the energy difference between the A and B absorption peaks of WS2. However, increasing the dielectric function on the surface of WS2 has a small influence on their optical properties. The PEG NP assembly on WS2 exhibited a significant red shift of the PL spectrum and a large decrease of the energy difference between A and B absorption peaks. Deforming the WS2 sheet by the PEG NP assembly decreased the orbital coupling and lowered the electronic direct band gap significantly. Raman bands of WS2 are shifted to a higher frequency on improving its mechanical strength after the PEG assembly.

10.
Am J Cancer Res ; 9(2): 347-362, 2019.
Article in English | MEDLINE | ID: mdl-30906633

ABSTRACT

Abnormal expression of dermatan sulfate epimerase (DSE) has been found in many types of cancer, while its expression and biological functions in hepatocellular carcinoma (HCC) progression remains obscure. Here we report that DSE, the enzyme that catalyzes the conversion of chondroitin sulfate (CS) to dermatan sulfate (DS), is a critical mediator of malignant character in HCC, through regulation of CCL5 signaling. DSE mRNA and protein were downregulated frequently in HCC tumors, where these events were associated with advanced tumor stages, metastases, and poor survival. DSE-mediated tumor growth was evaluated in immune-deficient and immune-complement mice models. Restoring DSE expression in HCC cells suppressed tumor growth, as well as decreased IL-1ß and CCL5 levels in transplanted tumor tissue. Mechanistic investigations revealed that the expression of DSE altered CCL5 signaling and cell surface binding in HCC cells. Accordingly, DSE suppressed CCL5-induced cell growth, migration, and invasion, whereas silencing of DSE enhanced CCL5-triggered malignant phenotypes. Inhibiting CCR1 activity with BX471 decreased CCL5-induced malignant characters caused by siRNA-mediated knockdown of DSE in HCC cells, establishing the critical role of the CCL5/CCR1 axis in mediating the effects of DSE expression. Taken together, our results suggest that DSE dysregulation contributes to the malignant behavior of HCC cells. This provides novel insight into the significance of DSE in CCL5 signaling and HCC pathogenesis.

11.
PLoS One ; 13(6): e0198364, 2018.
Article in English | MEDLINE | ID: mdl-29864158

ABSTRACT

Remodeling of the extracellular matrix (ECM) in the tumor microenvironment promotes glioma progression. Chondroitin sulfate (CS) proteoglycans appear in the ECM and on the cell surface, and can be catalyzed by dermatan sulfate epimerase to form chondroitin sulfate/dermatan sulfate (CS/DS) hybrid chains. Dermatan sulfate epimerase 1 (DSE) is overexpressed in many types of cancer, and CS/DS chains mediate several growth factor signals. However, the role of DSE in gliomas has never been explored. In the present study, we determined the expression of DSE in gliomas by consulting a public database and conducting immunohistochemistry on a tissue array. Our investigation revealed that DSE was upregulated in gliomas compared with normal brain tissue. Furthermore, high DSE expression was associated with advanced tumor grade and poor survival. We found high DSE expression in several glioblastoma cell lines, and DSE expression directly mediated DS chain formation in glioblastoma cells. Knockdown of DSE suppressed the proliferation, migration, and invasion of glioblastoma cells. In contrast, overexpression of DSE in GL261 cells enhanced these malignant phenotypes and in vivo tumor growth. Interestingly, we found that DSE selectively regulated heparin-binding EGF-like growth factor (HB-EGF)-induced signaling in glioblastoma cells. Inhibiting epidermal growth factor receptor (EGFR) and ErbB2 with afatinib suppressed DSE-enhanced malignant phenotypes, establishing the critical role of the ErbB pathway in regulating the effects of DSE expression. This evidence indicates that upregulation of DSE in gliomas contributes to malignant behavior in cancer cells. We provide novel insight into the significance of DS chains in ErbB signaling and glioma pathogenesis.


Subject(s)
Antigens, Neoplasm/metabolism , Brain Neoplasms/pathology , DNA-Binding Proteins/metabolism , Glioblastoma/pathology , Neoplasm Proteins/metabolism , Signal Transduction , Tissue Array Analysis/methods , Up-Regulation , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Male , Mice , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Transplantation , Phenotype , Survival Analysis
12.
Int J Biochem Cell Biol ; 86: 42-53, 2017 05.
Article in English | MEDLINE | ID: mdl-28323206

ABSTRACT

Astrocytes, a stellate-shape glial population in the central nervous system (CNS), maintain glutamate homeostasis in adult CNS by undergoing glutamate uptake at the synapse through their glutamate transporter-1 (GLT-1). Peroxisome proliferator-activated receptor-α (PPARα) can be activated by endogenous saturated fatty acids to regulate astrocytic lipid metabolism and functions. However, it is unclear if PPARα can exert the regulatory action on GLT-1 expression in astrocytes. This study showed that treatment with palmitic acid (PA) and the other two PPARα agonists (GW 7647 and WY 14,643) caused no change in the morphology of astrocytes, whereas membranous GLT-1 protein levels in astrocytes were significantly decreased by PA and PPARα agonists. Through lentivirus-mediated overexpression of GLT-1 tagged with red fluorescent protein (GLT-1-RFP), we also observed that GLT-1-RFP puncta in the processes of astrocytes were inhibited by the PPARα agonists. This reduction was prevented by the addition of the PPARα antagonist, GW6471. GLT-1-RFP was co-localized to the early endosome marker-EEA1 in astrocytes treated with the PPARα agonists. Moreover, PPARα-induced inhibition in membranous GLT-1 expression was abolished by the addition of dynamin inhibitor (dynasore). Furthermore, the co-treatment of astrocytes with PPARα agonists and dynasore, or with PPARα agonists and protein kinase C (PKC) inhibitor bis-indolylmaleimide 1 (BIS1), prevented the endocytosis of GLT-1-RFP. Based on the results, we conclude that the PPARα agonists increased GLT-1 endocytosis in astrocytes possibly through the PKC signaling pathway. In addition, our findings provide important information of PPARα involvement in the downregulation of astrocytic glutamate uptake via the promoted GLT-1 endocytosis.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Endocytosis/drug effects , Excitatory Amino Acid Transporter 2/metabolism , PPAR alpha/agonists , PPAR alpha/metabolism , Animals , Down-Regulation/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Glutamic Acid/metabolism , Ligands , Palmitic Acid/pharmacology , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/metabolism
13.
Stem Cells Transl Med ; 5(2): 235-47, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26718649

ABSTRACT

A major complication in continuous, ambulatory peritoneal dialysis in patients with end-stage renal disease who are undergoing long-term peritoneal dialysis (PD) is peritoneal fibrosis, which can result in peritoneal structural changes and functional ultrafiltration failure. Human umbilical mesenchymal stem cells (HUMSCs) in Wharton's jelly possess stem cell properties and are easily obtained and processed. This study focuses on the effects of HUMSCs on peritoneal fibrosis in in vitro and in vivo experiments. After 24-hour treatment with mixture of Dulbecco's modified Eagle's medium and PD solution at a 1:3 ratio, primary human peritoneal mesothelial cells became susceptible to PD-induced cell death. Such cytotoxic effects were prevented by coculturing with primary HUMSCs. In a rat model, intraperitoneal injections of 20 mM methylglyoxal (MGO) in PD solution for 3 weeks (the PD/MGO 3W group) markedly induced abdominal cocoon formation, peritoneal thickening, and collagen accumulation. Immunohistochemical analyses indicated neoangiogenesis and significant increase in the numbers of ED-1- and α-smooth muscle actin (α-SMA)-positive cells in the thickened peritoneum in the PD/MGO 3W group, suggesting that PD/MGO induced an inflammatory response. Furthermore, PD/MGO treatment for 3 weeks caused functional impairments in the peritoneal membrane. However, in comparison with the PD/MGO group, intraperitoneal administration of HUMSCs into the rats significantly ameliorated the PD/MGO-induced abdominal cocoon formation, peritoneal fibrosis, inflammation, neoangiogenesis, and ultrafiltration failure. After 3 weeks of transplantation, surviving HUMSCs were found in the peritoneum in the HUMSC-grafted rats. Thus, xenografts of HUMSCs might provide a potential therapeutic strategy in the prevention of peritoneal fibrosis. Significance: This study demonstrated that direct intraperitoneal transplantation of human umbilical mesenchymal stem cells into the rat effectively prevented peritoneal dialysis/methylglyoxal-induced abdominal cocoon formation, ultrafiltration failure, and peritoneal membrane alterations such as peritoneal thickening, fibrosis, and inflammation. These findings provide a basis for a novel approach for therapeutic benefits in the treatment of encapsulating peritoneal sclerosis.


Subject(s)
Graft Survival/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Neovascularization, Pathologic/prevention & control , Peritoneal Fibrosis/therapy , Wharton Jelly/cytology , Actins/genetics , Actins/metabolism , Animals , Biomarkers/metabolism , Cell Death , Culture Media/chemistry , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression , Humans , Injections, Intraperitoneal , Male , Mesenchymal Stem Cells/metabolism , Peritoneal Dialysis , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Peritoneum/metabolism , Peritoneum/pathology , Pyruvaldehyde , Rats , Rats, Sprague-Dawley , Transplantation, Heterologous , Umbilical Cord/cytology , Umbilical Cord/metabolism , Wharton Jelly/metabolism
14.
Mol Neurobiol ; 53(6): 3528-3539, 2016 08.
Article in English | MEDLINE | ID: mdl-26096706

ABSTRACT

B cell CLL/lymphoma 11B (Bcl11b), a C2H2 zinc finger transcription factor, not only serves as a critical regulator in development but also plays the controversial role in T cell acute lymphoblastic leukemia (T-ALL). We previously found that the enriched expression of Bcl11b was detected in high tumorigenic C6 glioma cells. However, the role of Bcl11b in glioma malignancy and its mechanisms remains to be uncovered. In this study, using the lentivirus-mediated knockdown (KD) approach, we found that Bcl11b KD in tumorigenic C6 cells reduced the cell proliferation, colony formation, and migratory ability. The results were further verified using two human malignant glioma cell lines, U87 and U251 cells. A cyclin-dependent kinase inhibitor p21, a known Bcl11b target, was significantly upregulated in tumorigenic C6, U87, and U251 cells after Bcl11b KD. Cellular senescence was observed by examination of the ß-galactosidase activity in U87 and U251 cells with Bcl11b KD. Reduced expression of stemness gene Sox-2 and its downstream effector Bmi-1 was also observed in U87 and U251 cells with Bcl11b KD. These results suggest that the ablation of Bcl11b gene expression induced glioma cell senescence. Propidium iodide (PI) staining combined with flow cytometry analysis also showed that Bcl11b KD led to the cell cycle arrest of U87 and U251 cells at the G0/G1 or at the S phase, indicating that Bcl11b is required for glioma cell cycle progression. Together, this is the first study to show that the inhibition of Bcl11b suppresses glioma cell growth by regulating the expression of the cell cycle regulator p21 and stemness-associated genes (Sox-2/Bmi-1).


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Brain Neoplasms/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/genetics , Gene Knockdown Techniques , Humans , Polycomb Repressive Complex 1/metabolism , Rats , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation/genetics
15.
PLoS One ; 10(5): e0126217, 2015.
Article in English | MEDLINE | ID: mdl-25938237

ABSTRACT

HYS-32 is a novel derivative of combretastatin-A4 (CA-4) previously shown to induce microtubule coiling in rat primary astrocytes. In this study, we further investigated the signaling mechanism and EB1, a microtubule-associated end binding protein, involved in HYS-32-induced microtubule catastrophes. Confocal microscopy with double immunofluorescence staining revealed that EB1 accumulates at the growing microtubule plus ends, where they exhibit a bright comet-like staining pattern in control astrocytes. HYS-32 induced microtubule catastrophes in both a dose- and time-dependent manner and dramatically increased the distances between microtubule tips and the cell border. Treatment of HYS-32 (5 µM) eliminated EB1 localization at the microtubule plus ends and resulted in an extensive redistribution of EB1 to the microtubule lattice without affecting the ß-tubulin or EB1 protein expression. Time-lapse experiments with immunoprecipitation further displayed that the association between EB-1 and ß-tubulin was significantly decreased following a short-term treatment (2 h), but gradually increased in a prolonged treatment (6-24 h) with HYS-32. Further, HYS-32 treatment induced GSK3ß phosphorylation at Y216 and S9, where the ratio of GSK3ß-pY216 to GSK3ß-pS9 was first elevated followed by a decrease over time. Co-treatment of astrocytes with HYS-32 and GSK3ß inhibitor SB415286 attenuated the HYS-32-induced microtubule catastrophes and partially prevented EB1 dissociation from the plus end of microtubules. Furthermore, co-treatment with PI3K inhibitor LY294002 inhibited HYS-32-induced GSK3ß-pS9 and partially restored EB1 distribution from the microtubule lattice to plus ends. Together these findings suggest that HYS-32 induces microtubule catastrophes by preventing EB1 from targeting to microtubule plus ends through the GSK3ß signaling pathway.


Subject(s)
4-Butyrolactone/analogs & derivatives , Astrocytes/drug effects , Astrocytes/metabolism , Glycogen Synthase Kinase 3/metabolism , Microtubules/metabolism , Naphthalenes/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , 4-Butyrolactone/pharmacology , Aminophenols/pharmacology , Animals , Cell Movement/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Maleimides/pharmacology , Neuroprotective Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Rats
16.
PLoS One ; 8(11): e79350, 2013.
Article in English | MEDLINE | ID: mdl-24236122

ABSTRACT

The astrocytic syncytium plays a critical role in maintaining the homeostasis of the brain through the regulation of gap junction intercellular communication (GJIC). Changes to GJIC in response to inflammatory stimuli in astrocytes may have serious effects on the brain. We have previously shown that lipopolysaccharide (LPS) reduces connexin43 (Cx43) expression and GJIC in cultured rat astrocytes via a toll-like receptor 4-mediated signaling pathway. In the present study, treatment of astrocytes with LPS resulted in a significant increase in levels of the phosphorylated forms of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) -1, -2, and -3 for up to 18 h. An increase in nuclear transcription factor NF-κB levels was also observed after 8 h of LPS treatment and was sustained for up to 18 h. The LPS-induced decrease in Cx43 protein levels and inhibition of GJIC were blocked by the SAPK/JNK inhibitor SP600125, but not by the NF-κB inhibitor BAY11-7082. Following blockade of de novo protein synthesis by cycloheximide, LPS accelerated Cx43 degradation. Moreover, the LPS-induced downregulation of Cx43 was blocked following inhibition of 26S proteasome activity using the reversible proteasome inhibitor MG132 or the irreversible proteasome inhibitor lactacystin. Immunoprecipitation analyses revealed an increased association of Cx43 with both ubiquitin and E3 ubiquitin ligase Nedd4 in astrocytes after LPS stimulation for 6 h and this effect was prevented by SP600125. Taken together, these results suggest that LPS stimulation leads to downregulation of Cx43 expression and GJIC in rat astrocytes by activation of SAPK/JNK and the ubiquitin-proteasome proteolytic pathway.


Subject(s)
Astrocytes/metabolism , Connexin 43/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitins/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Anthracenes/pharmacology , Astrocytes/drug effects , Connexin 43/genetics , Down-Regulation/drug effects , Gap Junctions/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Leupeptins/pharmacology , Lipopolysaccharides , NF-kappa B/metabolism , Proteolysis , Rats , Ubiquitination
17.
Toxicology ; 314(1): 11-21, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-23973256

ABSTRACT

Lysophosphatidylcholine (LPC) is a potent pro-arrhythmic derivative of the membrane phosphotidylcholine, which is accumulated in heart tissues during cardiac ischemia. However, the cellular mechanism underlying LPC-induced cardiomyocyte damage remains to be elucidated. This study focuses on the effects of LPC on cardiomyocyte gap junction. At 30µM, LPC decreased the spontaneous contraction rates of cardiomyocytes, and caused arrhythmic contraction without affecting cell viability. Connexin43 (Cx43) was seen as large plaques at cell junctions in control cells, whereas upon LPC treatment, the intensity of Cx43 staining was decreased in a concentration-sensitive manner and Cx43 staining appeared as tiny dots at cell junctions with a corresponding increase in cytoplasmic punctate staining. This distributional change of Cx43 was accompanied by an impairment of the gap junction intercellular communication (GJIC). Further, LPC treatment induced protein kinase C (PKC) activation, and PKC-dependent Cx43 phosphorylation at serine (Ser) 368. Pre-treatment with a specific PKCɛ inhibitor, eV1-2, prevented the LPC-induced Cx43 phosphorylation at Ser368 and the loss of Cx43 from gap junctions, both of which may disturb GJIC functions. Furthermore, siRNA knockdown of PKCɛ in H9c2 cells prevented LPC-induced serine phosphorylation of Cx43, confirming the role of PKCɛ in Cx43 serine phosphorylation. Double labeling immunofluorescence showed that LPC increased the colocalization of Cx43 with ubiquitin, and pretreatment with MG132 effectively prevented LPC-induced gap junction disassembly. LPC increased the ubiquitination of Cx43, which was blocked by eV1-2 pretreatment, suggesting that LPC accelerated the intracellular degradation of Cx43 via the ubiquitin-proteasomal pathway. It can be concluded that LPC destroyed the structure and function of gap junctions via PKCɛ-mediated serine phosphorylation of Cx43. PKCɛ inhibitors might therefore be effective in prevention of LPC-related diseases.


Subject(s)
Connexin 43/metabolism , Lysophosphatidylcholines/pharmacology , Myocytes, Cardiac/metabolism , Protein Kinase C-epsilon/metabolism , Serine/metabolism , Animals , Animals, Newborn , Blotting, Western , Cell Survival/drug effects , Coloring Agents , Female , Fluorescent Antibody Technique , Heart Rate/drug effects , Immunoprecipitation , Indoles , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Phosphorylation , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Tetrazolium Salts , Thiazoles
18.
Neurochem Int ; 62(6): 881-92, 2013 May.
Article in English | MEDLINE | ID: mdl-23500605

ABSTRACT

HYS-32 [4-(3,4-dimethoxyphenyl)-3-(naphthalen-2-yl)-2(5H)-furanone] is a new analogue of the anti-tumor compound combretastatin A-4 containing a cis-stilbene moiety. In this study, we investigated its effects on Cx43 gap junction intercellular communication (GJIC) and the signaling pathway involved in rat primary astrocytes. Western blot analyses showed that HYS-32 dose- and time-dependently upregulated Cx43 expression. A confocal microscopic study and scrape-loading/dye transfer analyses demonstrated that HYS-32 (5µM) induced microtubule coiling, accumulation of Cx43 in gap junction plaques, and increased GJIC in astrocytes. The HYS-32-induced microtubule coiling and Cx43 accumulation in gap junction plaques was reversed when HYS-32 was removed. Treatment of astrocytes with cycloheximide resulted in time-dependent degradation of by co-treatment with HYS-32 by increasing the half-life of Cx43. Co-treatment with HYS-32 also prevented the LPS-induced downregulation of Cx43 and inhibition of GJIC in astrocytes. HYS-32 induced activation of PKC, ERK, and JNK, and co-treatment with the PKC inhibitor Go6976 or the ERK inhibitor PD98059, but not the JNK inhibitor SP600125, prevented the HYS-32-induced increase in Cx43 expression and GJIC. Go6976 suppressed the HYS-32-induced PKC phosphorylation and increase in phospho-ERK levels, while PD98059 did not prevent the HYS-32-induced increase in phospho-PKC levels, suggesting that PKC is an upstream effector of ERK. In conclusion, our results show that HYS-32 increases the half-life of Cx43 and enhances Cx43 expression and GJIC in astrocytes via a PKC-ERK signaling cascade. These novel biological effects of HYS-32 on astrocyte gap junctions support its potential for therapeutic use as a protective agent for the central nervous system.


Subject(s)
4-Butyrolactone/analogs & derivatives , Antineoplastic Agents, Phytogenic/pharmacology , Astrocytes/drug effects , Cell Communication/drug effects , Connexin 43/biosynthesis , Gap Junctions/drug effects , Naphthalenes/pharmacology , Stilbenes/pharmacology , 4-Butyrolactone/pharmacology , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Coloring Agents , Enzyme Inhibitors/pharmacology , Female , Image Processing, Computer-Assisted , Male , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Tetrazolium Salts , Thiazoles
19.
Neurotoxicology ; 35: 169-79, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23403203

ABSTRACT

Antofine, a phenanthroindolizidine alkaloid derived from Cryptocaryachinensis and Ficusseptica in the Asclepiadaceae milkweed family, is cytotoxic for various cancer cell lines. In this study, we demonstrated that treatment of rat primary astrocytes with antofine induced dose-dependent inhibition of gap junction intercellular communication (GJIC), as assessed by scrape-loading 6-carboxyfluorescein dye transfer. Levels of Cx43 protein were also decreased in a dose- and time-dependent manner following antofine treatment. Double-labeling immunofluorescence microscopy showed that antofine (10ng/ml) induced endocytosis of surface gap junctions into the cytoplasm, where Cx43 was co-localized with the early endosome marker EEA1. Inhibition of lysosomes or proteasomes by co-treatment with antofine and their respective specific inhibitors, NH4Cl or MG132, partially inhibited the antofine-induced decrease in Cx43 protein levels, but did not inhibit the antofine-induced inhibition of GJIC. After 30min of treatment, antofine induced a rapid increase in the intracellular Ca(2+) concentration and activation of protein kinase C (PKC)α/ßII, which was maintained for at least 6h. Co-treatment of astrocytes with antofine and the intracellular Ca(2+) chelator BAPTA-AM prevented downregulation of Cx43 and inhibition of GJIC. Moreover, co-treatment with antofine and a specific PKCß inhibitor prevented endocytosis of gap junctions, downregulation of Cx43, and inhibition of GJIC. Taken together, these findings indicate that antofine induces Cx43 gap junction disassembly by the PKCß signaling pathway. Inhibition of GJIC by antofine may undermine the neuroprotective effect of astrocytes in CNS.


Subject(s)
Astrocytes/drug effects , Cell Communication/drug effects , Connexin 43/metabolism , Gap Junctions/drug effects , Indoles/toxicity , Phenanthrolines/toxicity , Protein Kinase C/metabolism , Animals , Animals, Newborn , Astrocytes/enzymology , Astrocytes/pathology , Calcium/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Endocytosis/drug effects , Enzyme Activation , Female , Gap Junctions/enzymology , Gap Junctions/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Male , Microscopy, Fluorescence , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Vesicular Transport Proteins/metabolism
20.
Cell Biol Int ; 36(9): 833-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22582758

ABSTRACT

LPA (lysophosphatidic acid) is a natural phospholipid that plays important roles in promoting cancer cell proliferation, invasion and metastases. We previously reported that LPA induces ovarian cancer cell dispersal and disruption of AJ (adherens junction) through the activation of SFK (Src family kinases). In this study, we have investigated the regulatory mechanisms during the early phase of LPA-induced cell dispersal. An in vitro model of the ovarian cancer cell line SKOV3 for cell dispersal was used. LPA induces rapid AJ disruption by increasing the internalization of N-cadherin-ß-catenin. By using immunoprecipitations, LPA was shown to induce increased tyrosine phosphorylation of ß-catenin and alter the balance of ß-catenin-bound SFK and PTP1B (phosphotyrosine phosphatase 1B). The altered balance of tyrosine kinase/phosphatase correlated with a concomitant disintegration of the ß-catenin-α-catenin, but not the ß-catenin-N-cadherin complex. This disintegration of ß-catenin from α-catenin and the cell dispersal caused by LPA can be rescued by blocking SFK activity with the chemical inhibitor, PP2. More importantly, PP2 also restores the level of PTP1B bound to ß-catenin. We propose that LPA signalling alters AJ stability by changing the dynamics of tyrosine kinase/phosphatase bound to AJ proteins. This work provides further understanding of the early signalling events regulating ovarian cancer cell dispersal and AJ disruption induced by LPA.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Catenins/metabolism , Lysophospholipids/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Multiprotein Complexes/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Ovarian Neoplasms , Phosphorylation , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL