Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 12(1): 2921, 2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35190601

ABSTRACT

Inorganic metal oxides with the merits of high carrier transport capability, low cost and superior chemical stability have largely served as the hole transport layer (HTL) in perovskite solar cells (PSCs) in recent years. Among them, ternary metal oxides have gradually attracted attention because of the wide tenability of the two inequivalent cations in the lattice sites that offer interesting physicochemical properties. In this work, ZnCo2O4 nanoparticles (NPs) were prepared by a chemical precipitation method and served as the HTL in inverted PSCs. The device based on the ZnCo2O4 NPs HTL showed better efficiency of 12.31% and negligible hysteresis compared with the one using PEDOT:PSS film as the HTL. Moreover, the device sustained 85% of its initial efficiency after 240 h storage under a halogen lamps matrix exposure with an illumination intensity of 1000 W/m2, providing a powerful strategy to design long term stable PSCs for future production.

2.
Sci Rep ; 6: 25082, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27122225

ABSTRACT

Statins are used widely to lower serum cholesterol and the incidence of cardiovascular diseases. Growing evidence shows that statins also exhibit beneficial effects against cancers. In this study, we investigated the molecular mechanisms involved in lovastatin-induced cell death in Fadu hypopharyngeal carcinoma cells. Lovastatin caused cell cycle arrest and apoptosis in FaDu cells. Lovastatin increased p21(cip/Waf1) level while the survivin level was decreased in the presence of lovastatin. Survivin siRNA reduced cell viability and induced cell apoptosis in FaDu cells. Lovastatin induced phosphorylation of AMP-activated protein kinase (AMPK), p38 mitogen-activated protein kinase (MAPK) and transcription factor p63. Lovastatin also caused p63 acetylation and increased p63 binding to survivin promoter region in FaDu cells. AMPK-p38MAPK signaling blockade abrogated lovastatin-induced p63 phosphorylation. Lovastatin's enhancing effect on p63 acetylation was reduced in HDAC3- or HDAC4- transfected cells. Moreover, transfection of cells with AMPK dominant negative mutant (AMPK-DN), HDAC3, HDAC4 or p63 siRNA significantly reduced lovastatin's effects on p21(cip/Waf1) and survivin. Furthermore, lovastatin inhibited subcutaneous FaDu xenografts growth in vivo. Taken together, lovastatin may activate AMPK-p38MAPK-p63-survivin cascade to cause FaDu cell death. This study establishes, at least in part, the signaling cascade by which lovastatin induces hypopharyngeal carcinoma cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death , Epithelial Cells/drug effects , Lovastatin/pharmacology , AMP-Activated Protein Kinases/metabolism , Cell Line, Tumor , Humans , Inhibitor of Apoptosis Proteins/metabolism , Signal Transduction , Survivin , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
3.
Oncotarget ; 6(1): 85-100, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25415226

ABSTRACT

Angiogenesis, one of the major routes for tumor invasion and metastasis represents a rational target for therapeutic intervention. Recent development in drug discovery has highlighted the diverse biological and pharmacological properties of hydroxamate derivatives. In this study, we characterized the anti-angiogenic activities of a novel aliphatic hydroxamate, WMJ-S-001, in an effort to develop novel angiogenesis inhibitors. WMJ-S-001 inhibited vascular endothelial growth factor (VEGF)-A-induced proliferation, invasion and endothelial tube formation of human umbilical endothelial cells (HUVECs). WMJ-S-001 suppressed VEGF-A-induced microvessel sprouting from aortic rings, and attenuated angiogenesis in in vivo mouse xenograft models. In addition, WMJ-S-001 inhibited the phosphorylations of VEGFR2, Src, FAK, Akt and ERK in VEGF-A-stimulated HUVECs. WMJ-S-001 caused an increase in SHP-1 phosphatase activity, whereas NSC-87877, a SHP-1 inhibitor, restored WMJ-S-001 suppression of VEGFR2 phosphorylation and cell proliferation. Furthermore, WMJ-S-001 inhibited cell cycle progression and induced cell apoptosis in HUVECs. These results are associated with p53 phosphorylation and acetylation and the modulation of p21 and survivin. Taken together, WMJ-S-001 was shown to modulate vascular endothelial cell remodeling through inhibiting VEGFR2 signaling and induction of apoptosis. These results also support the role of WMJ-S-001 as a potential drug candidate and warrant the clinical development in the treatment of cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Naphthalenes/pharmacology , Neoplasms/drug therapy , Neovascularization, Pathologic , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Apoptosis , Cell Cycle , Cell Proliferation , Cell Survival , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Human Umbilical Vein Endothelial Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Nude , Microcirculation , Neoplasm Transplantation , Neoplasms/metabolism , Phosphorylation , Protein Structure, Tertiary , Signal Transduction , Survivin
4.
Angiogenesis ; 17(2): 407-18, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24048742

ABSTRACT

Formation of lymphatic capillaries by lymphatic endothelial cells (LECs) occurs both in normal tissues as well as in pathological processes including tumor metastasis. Interleukin-6 (IL-6), a potent pro-inflammatory cytokine, has been shown to be highly elevated in various cancers. IL-6 has also been shown to increase tumor lymphangiogenesis through vascular endothelial growth factor-C (VEGF-C) induction in tumor cells. Although lymphangiogenesis is associated with lymph node metastasis and also resistance to conventional therapy in various cancers, the precise mechanisms of lymphangiogenesis in LECs remain unclear. This study aimed to investigate the signaling cascade involved in IL-6-induced VEGF-C expression in murine LECs (SV-LEC). The VEGF-C mRNA and protein levels were increased in SV-LECs exposed to IL-6. IL-6 time-dependently induced Src phosphorylation and downstream phosphorylation of ERK1/2 and p38MAPK. In contrast, PP2, an inhibitor of Src signaling, abrogated IL-6's effects on ERK1/2 and p38MAPK phosphorylation. IL-6 exposure also led to increase in VEGF-C promoter-luciferase activity as well as C/EBPß- and κB-luciferase activities. VEGF-C promoter-, C/EBPß- and κB-luciferase activities were all suppressed by Src, ERK1/2 or p38MAPK signaling blockades despite presence of IL-6. Finally, C/EBPß and p65 binding to the VEGF-C promoter region were increased after IL-6 exposure in SV-LECs. Taken together, we report a Src-mediated ERK1/2 and p38MAPK activation resulting in C/EBPß and p65 binding to the promoter region of VEGF-C, leading to VEGF-C expression in IL-6-exposed SV-LECs.


Subject(s)
Endothelial Cells/metabolism , Interleukin-6/pharmacology , Vascular Endothelial Growth Factor C/metabolism , src-Family Kinases/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , NF-kappa B/metabolism , Phosphorylation/drug effects , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
PLoS One ; 8(11): e81592, 2013.
Article in English | MEDLINE | ID: mdl-24312323

ABSTRACT

Angiogenesis occurs during tissue growth, development and wound healing. It is also required for tumor progression and represents a rational target for therapeutic intervention. NBM-T-BMX-OS01 (BMX), derived from the semisynthesis of osthole, an active ingredient isolated from Chinese herb Cnidium monnieri (L.) Cuss., was recently shown to enhance learning and memory in rats. In this study, we characterized the anti-angiogenic activities of NBM-T-BMX-OS01 (BMX) in an effort to develop novel inhibitors to suppress angiogenesis and tumor growth. BMX inhibited vascular endothelial growth factor (VEGF)-induced proliferation, migration and endothelial tube formation in human umbilical endothelial cells (HUVECs). BMX also attenuated VEGF-induced microvessel sprouting from aortic rings ex vivo and reduced HCT116 colorectal cancer cells-induced angiogenesis in vivo. Moreover, BMX inhibited the phosphorylation of VEGFR2, FAK, Akt and ERK in HUVECs exposed to VEGF. BMX was also shown to inhibit HCT116 cell proliferation and to suppress the growth of subcutaneous xenografts of HCT116 cells in vivo. Taken together, this study provides evidence that BMX modulates vascular endothelial cell remodeling and leads to the inhibition of tumor angiogenesis. These results also support the role of BMX as a potential drug candidate and warrant the clinical development in the treatment of cancer.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Coumarins/chemistry , Coumarins/pharmacology , Neovascularization, Pathologic/drug therapy , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Capillaries/drug effects , Capillaries/metabolism , Capillaries/physiology , Cell Movement/drug effects , Cell Proliferation/drug effects , Coumarins/therapeutic use , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
6.
Biochim Biophys Acta ; 1830(8): 4053-64, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23583370

ABSTRACT

BACKGROUND: Statins, the 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors with cholesterol-lowering properties, were recently shown to exhibit anti-cancer effects. However, the molecular mechanism underlying statin-induced cancer cell death remains to be elucidated. Elevated level of survivin is often found over-expressed in human cancers and has been implicated in the progression of tumorigenesis. Given its central role in cell division and action as an apoptosis suppressor, survivin represents a potential molecular target in cancer management. METHODS: In this study, we explored the underlying mechanisms in simvastatin-induced HCT116 colorectal cancer cell apoptosis. RESULTS: Simvastatin decreased cell viability and induced cell apoptosis in HCT116 cells. These results are associated with the modulation of p21(cip/Waf1) and survivin. Survivin knockdown using survivin siRNAs also decreased cell viability and induced cell apoptosis. Simvastatin's actions on p21(cip/Waf1), survivin and apoptosis were reduced in p53 null HCT116 cells. Simvastatin caused an increase in p53 phosphorylation and acetylation. In addition, simvastatin activated p38 mitogen-activated protein kinase (p38MAPK), whereas an inhibitor of p38MAPK signaling abrogated simvastatin's effects of increasing p53 and p21(cip/Waf1) promoter luciferase activity. Cell viability and survivin promoter luciferase activity in the presence of simvastatin were also restored by p38MAPK inhibitor. Furthermore, Sp1 binding to the survivin promoter region decreased while p53 and p63 binding to the promoter region increased after simvastatin exposure. CONCLUSIONS: Simvastatin activates the p38MAPK-p53-survivin cascade to cause HCT116 colorectal cancer cell apoptosis. GENERAL SIGNIFICANCE: This study delineates, in part, the underlying mechanisms of simvastatin in decreasing survivin and subsequent colorectal cancer cell apoptosis.


Subject(s)
Apoptosis/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inhibitor of Apoptosis Proteins/physiology , Signal Transduction , Simvastatin/pharmacology , Tumor Suppressor Protein p53/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Acetylation , Cell Survival/drug effects , HCT116 Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Promoter Regions, Genetic , Survivin , Xenograft Model Antitumor Assays
7.
Biochim Biophys Acta ; 1820(2): 104-15, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22155142

ABSTRACT

BACKGROUND: Elevated levels of survivin and histone deacetylases (HDACs) are often found over-expressed in human cancers, including colorectal cancer, and have been implicated in tumorigenesis. HDAC inhibition induces growth arrest and cell death in various transformed cell; however, the mechanisms by which this reduces cell viability in colorectal cancer cells remain unexplained. METHODS: We explored the actions of two HDAC inhibitors, trichostatin A (TSA) and sirtinol, in HT29 colon cancer cells. RESULTS: TSA and sirtinol induced apoptosis and inhibited cell proliferation in HT29 cells. These results are associated with the modulation of survivin. Survivin promoter luciferase activity and Sp1, a transcription factor that contributes to survivin expression, were suppressed in cells exposed to TSA or sirtinol. TSA and sirtinol also activated p38 mitogen-activated protein kinase (p38MAPK) and AMP-activated protein kinase (AMPK). Inhibitors of p38MAPK or AMPK signaling abrogated TSA and sirtinol's effects of decreasing cell viability. Survivin promoter luciferase activity in the presence of TSA or sirtinol was restored by AMPK dominant negative mutant or p38MAPK inhibitor. Furthermore, Sp1 binding to the survivin promoter region decreased while p63 binding to the promoter region increased after TSA or sirtinol exposure. CONCLUSIONS: We report a p38MAPK- and AMPK-mediated downregulation of survivin, and its functional correlation with decreased colon cancer cell viability in the presence of HDAC inhibitor. p63 and Sp1 may also contribute to TSA and sirtinol actions. GENERAL SIGNIFICANCE: This study delineates, in part, the underlying mechanisms of TSA and sirtinol in decreasing survivin expression and subsequent colon cancer cell viability.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Benzamides/pharmacology , Colonic Neoplasms/enzymology , Hydroxamic Acids/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Naphthols/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Luciferases/metabolism , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Sp1 Transcription Factor/metabolism , Survivin , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
8.
Biochim Biophys Acta ; 1810(12): 1160-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21911040

ABSTRACT

BACKGROUND: Histone deacetylase (HDAC) inhibitors have emerged as a new class of antitumor agents because they were demonstrated to induce cell cycle arrest, promote cell apoptosis, and inhibit metastasis. Recently, HDAC inhibitors were also shown to exhibit pronounced anti-inflammatory properties. However, the underlying mechanism contributing to the suppression of inflammatory responses by HDAC inhibitors remains to be fully defined. In the present study, we explored the actions of trichostatin A (TSA), a potent HDAC inhibitor, on lipopolysaccharide (LPS)-induced cyclooxygenase (COX)-2 expression in human umbilical vascular endothelial cells (HUVECs). METHODS: HUVECs were exposed to LPS in the absence or presence of TSA. COX-2 expression and signaling molecules (JNK, p38MAPK and c-jun) activated by LPS were assessed. RESULTS: The LPS-induced cox-2 messenger RNA and protein were markedly suppressed by TSA. TSA inhibited JNK and p38MAPK phosphorylation in cells exposed to LPS. Treatment of cells with a JNK signaling inhibitor (JNK inhibitor II) or a p38MAPK inhibitor (p38MAPK inhibitor III) markedly inhibited LPS-induced COX-2 expression. TSA suppression of JNK and p38MAPK phosphorylation and subsequent COX-2 expression were restored by selective inhibition of MKP-1 using MKP-1 siRNA. In addition, TSA caused an increase in MKP-1 phosphatase activity in HUVECs. In conclusion, TSA may cause MKP-1 activation to dephosphorylate JNK and p38MAPK, leading to the downregulation of COX-2 in HUVECs stimulated by LPS, a proinflammatory stimulus. GENERAL SIGNIFICANCE: MKP-1 contributes to TSA's protective actions in HUVECs exposed to LPS. The present study also supports the therapeutic value of TSA in treating inflammatory vascular diseases.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/drug effects , Dual Specificity Phosphatase 1/metabolism , Endothelium, Vascular/drug effects , Hydroxamic Acids/pharmacology , Umbilical Veins/drug effects , Base Sequence , Blotting, Western , Cells, Cultured , Cyclooxygenase 2/metabolism , DNA Primers , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Humans , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins/cytology , Umbilical Veins/enzymology
9.
Biochim Biophys Acta ; 1810(5): 504-13, 2011 May.
Article in English | MEDLINE | ID: mdl-21376104

ABSTRACT

BACKGROUND: Histone deacetylase (HDAC) inhibitors were demonstrated to induce cell cycle arrest, promote cell differentiation or apoptosis, and inhibit metastasis. HDAC inhibitors have thus emerged as a new class of anti-tumor agents for various types of tumors. However, the mechanisms by which HDAC inhibition-induced cell death remain to be fully defined. METHODS: In the present study, we explored the apoptotic actions of trichostatin A (TSA), a HDAC inhibitor, in C6 glioma cells. RESULTS: TSA activated p38 mitogen-activated protein kinase (p38MAPK), leading to p53 phosphorylation and activation. P53, a proapoptotic transcription factor, in turn transactivated the expression of a proapoptotic protein, Bax. In addition, survivin, a member of inhibitor of apoptotic protein, was significantly decreased in TSA-treated C6 cells. P53 recruited to the endogenous survivin promoter region was increased and accompanied by decreasing recruitment of SP1 in response to TSA. TSA was also shown to induce IKK dephosphorylation and to suppress NF-κB reporter activity. CONCLUSIONS: TSA may cause C6 cell apoptosis through activating p38MAPK-p53 cascade resulting in Bax expression and survivin suppression. Negative regulation of IKK-NF-κB signaling may also lead to p53 activation and contribute to TSA apoptotic actions. GENERAL SIGNIFICANCE: TSA-induced p53 activation may occur through p53 modification by phosphorylation or by acetylation via IKK inactivation. The present study delineates, in part, the signaling pathways involved in TSA-induced glioma cell death.


Subject(s)
Apoptosis/drug effects , Hydroxamic Acids/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , Dose-Response Relationship, Drug , Flow Cytometry , Glioma/metabolism , Glioma/pathology , Histone Deacetylase Inhibitors/pharmacology , I-kappa B Kinase/metabolism , Microtubule-Associated Proteins/genetics , Models, Biological , NF-kappa B/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding , Rats , Survivin , Time Factors , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...