Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Neuroimmune Pharmacol ; 19(1): 11, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530514

ABSTRACT

Neuro-inflammation involves distinct alterations of microglial phenotypes, containing nocuous pro-inflammatory M1-phenotype and neuroprotective anti-inflammatory M-phenotype. Currently, there is no effective treatment for modulating such alterations. M1/M2 marker of primary microglia influenced by Melatonin were detected via qPCR. Functional activities were explored by western blotting, luciferase activity, EMSA, and ChIP assay. Structure interaction was assessed by molecular docking and LIGPLOT analysis. ER-stress detection was examined by ultrastructure TEM, calapin activity, and ERSE assay. The functional neurobehavioral evaluations were used for investigation of Melatonin on the neuroinflammation in vivo. Melatonin had targeted on Peroxisome Proliferator Activated Receptor Delta (PPARδ) activity, boosted LPS-stimulated alterations in polarization from the M1 to the M2 phenotype, and thereby inhibited NFκB-IKKß activation in primary microglia. The PPARδ agonist L-165,041 or over-expression of PPARδ plasmid (ov-PPARδ) showed similar results. Molecular docking screening, dynamic simulation approaches, and biological studies of Melatonin showed that the activated site was located at PPARδ (phospho-Thr256-PPARδ). Activated microglia had lowered PPARδ activity as well as the downstream SIRT1 formation via enhancing ER-stress. Melatonin, PPARδ agonist and ov-PPARδ all effectively reversed the above-mentioned effects. Melatonin blocked ER-stress by regulating calapin activity and expression in LPS-activated microglia. Additionally, Melatonin or L-165,041 ameliorated the neurobehavioral deficits in LPS-aggravated neuroinflammatory mice through blocking microglia activities, and also promoted phenotype changes to M2-predominant microglia. Melatonin suppressed neuro-inflammation in vitro and in vivo by tuning microglial activation through the ER-stress-dependent PPARδ/SIRT1 signaling cascade. This treatment strategy is an encouraging pharmacological approach for the remedy of neuro-inflammation associated disorders.


Subject(s)
Melatonin , PPAR delta , Rats , Mice , Animals , Microglia , PPAR delta/metabolism , PPAR delta/pharmacology , PPAR delta/therapeutic use , Melatonin/pharmacology , Lipopolysaccharides/pharmacology , Sirtuin 1/metabolism , Molecular Docking Simulation , Inflammation/metabolism
2.
Cell Biol Toxicol ; 39(5): 1873-1896, 2023 10.
Article in English | MEDLINE | ID: mdl-34973135

ABSTRACT

BACKGROUND AND PURPOSE: Histone deacetylase (HDAC) inhibitors (HDIs) can modulate the epithelial-mesenchymal transition (EMT) progression and inhibit the migration and invasion of cancer cells. Emerging as a novel class of anti-cancer drugs, HDIs are attracted much attention in the field of drug discovery. This study aimed to discern the underlying mechanisms of Honokiol in preventing the metastatic dissemination of gastric cancer cells by inhibiting HDAC3 activity/expression. EXPERIMENTAL APPROACH: Clinical pathological analysis was performed to determine the relationship between HDAC3 and tumor progression. The effects of Honokiol on pharmacological characterization, functional, transcriptional activities, organelle structure changes, and molecular signaling were analyzed using binding assays, differential scanning calorimetry, luciferase reporter assay, HDAC3 activity, ER stress response element activity, transmission electron microscopy, immune-blotting, and Wnt/ß-catenin activity assays. The in vivo effects of Honokiol on peritoneal dissemination were determined by a mouse model and detected by PET/CT tomography. KEY RESULTS: HDAC3 over-expression was correlated with poor prognosis. Honokiol significantly abolished HDAC3 activity (Y298) via inhibition of NFκBp65/CEBPß signaling, which could be reversed by the over-expression of plasmids of NFκBp65/CEBPß. Treatments with 4-phenylbutyric acid (a chemical chaperone) and calpain-2 gene silencing inhibited Honokiol-inhibited NFκBp65/CEBPß activation. Honokiol increased ER stress markers and inhibited EMT-associated epithelial markers, but decreased Wnt/ß-catenin activity. Suppression of HDAC3 by both Honokiol and HDAC3 gene silencing decreased cell migration and invasion in vitro and metastasis in vivo. CONCLUSIONS AND IMPLICATIONS: Honokiol acts by suppressing HDAC3-mediated EMT and metastatic signaling. By prohibiting HDAC3, metastatic dissemination of gastric cancer may be blocked. Conceptual model showing the working hypothesis on the interaction among Honokiol, HDAC3, and ER stress in the peritoneal dissemination of gastric cancer. Honokiol targeting HDAC3 by ER stress cascade and mitigating the peritoneal spread of gastric cancer. Honokiol-induced ER stress-activated calpain activity targeted HDAC3 and blocked Tyr298 phosphorylation, subsequently blocked cooperating with EMT transcription factors and cancer progression. The present study provides evidence to demonstrate that HDAC3 is a positive regulator of EMT and metastatic growth of gastric cancer cells. The findings here imply that overexpressed HDAC3 is a potential therapeutic target for honokiol to reverse EMT and prevent gastric cancer migration, invasion, and metastatic dissemination. • Honokiol significantly abolished HDAC3 activity on catalytic tyrosine 298 residue site. In addition, Honokiol-induced ER stress markedly inhibited HDAC3 expression via inhibition of NFκBp65/CEBPß signaling. • HDAC3, which is a positive regulator of metastatic gastric cancer cell growth, can be significantly inhibited by Honokiol. • Opportunities for HDAC3 inhibition may be a potential therapeutic target for preventing gastric cancer metastatic dissemination.


Subject(s)
Stomach Neoplasms , beta Catenin , Animals , Mice , Calpain/antagonists & inhibitors , Calpain/genetics , Calpain/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Histone Deacetylases/metabolism , Positron Emission Tomography Computed Tomography , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Histone Deacetylase Inhibitors
3.
Biomed Pharmacother ; 155: 113725, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36152407

ABSTRACT

Diabetic retinopathy (DR) is a pathophysiologic vasculopathic process with obscure mechanisms and limited effective therapeutic strategies. Aryl hydrocarbon receptor (AhR) is an important regulator of xenobiotic metabolism and an environmental sensor. The aim of the present study was to investigate the role of AhR in the development of DR and elucidate the molecular mechanism of its downregulation. DR was evaluated in diabetes-induced retinal injury in wild type and AhR knockout (AhR-/-) mice. Retinal expression of AhR was determined in human donor and mice eyes by immunofluorescence since AhR activity was examined in diabetes. AhR knockout (AhRKO) mice were used to induce diabetes with streptozotocin, high-fat diet, or genetic double knockout with diabetes spontaneous mutation (Leprdb) (DKO; AhR-/-×Leprdb/db) for investigating structural, functional, and metabolic abnormalities in vascular and epithelial retina. Structural molecular docking simulation was used to survey the pharmacologic AhR agonists targeting phosphorylated AhR (Tyr245). Compared to diabetic control mice, diabetic AhRKO mice had aggravated alterations in retinal vasculature that amplified hallmark features of DR like vasopermeability, vascular leakage, inflammation, blood-retinal barrier breakdown, capillary degeneration, and neovascularization. AhR agonists effectively inhibited inflammasome formation and promoted AhR activity in human retinal microvascular endothelial cells and pigment epithelial cells. AhR activity and protein expression was downregulated, resulting in a decrease in DNA promoter binding site of pigment epithelium-derived factor (PEDF) by gene regulation in transcriptional cascade. This was reversed by AhR agonists. Our study identified a novel of DR model that target the protective AhR/PEDF axis can potentially maintain retinal vascular homeostasis, providing opportunities to delay the development of DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Mice , Humans , Animals , Diabetic Retinopathy/drug therapy , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Streptozocin/pharmacology , Endothelial Cells/metabolism , Inflammasomes/metabolism , Molecular Docking Simulation , Xenobiotics/metabolism , Retina , Mice, Inbred C57BL , Diabetes Mellitus/metabolism
4.
Arterioscler Thromb Vasc Biol ; 41(1): e46-e62, 2021 01.
Article in English | MEDLINE | ID: mdl-33176446

ABSTRACT

OBJECTIVE: Diabetic retinopathy, one of retinal vasculopathy, is characterized by retinal inflammation, vascular leakage, blood-retinal barrier breakdown, and neovascularization. However, the molecular mechanisms that contribute to diabetic retinopathy progression remain unclear. Approach and Results: Tpl2 (tumor progression locus 2) is a protein kinase implicated in inflammation and pathological vascular angiogenesis. Nε-carboxymethyllysine (CML) and inflammatory cytokines levels in human sera and in several diabetic murine models were detected by ELISA, whereas liquid chromatography-tandem mass spectrometry analysis was used for whole eye tissues. The CML and p-Tpl2 expressions on the human retinal pigment epithelium (RPE) cells were determined by immunofluorescence. Intravitreal injection of pharmacological inhibitor or NA (neutralizing antibody) was used in a diabetic rat model. Retinal leukostasis, optical coherence tomography, and H&E staining were used to observe pathological features. Sera of diabetic retinopathy patients had significantly increased CML levels that positively correlated with diabetic retinopathy severity and foveal thickness. CML and p-Tpl2 expressions also significantly increased in the RPE of both T1DM and T2DM diabetes animal models. Mechanistic studies on RPE revealed that CML-induced Tpl2 activation and NADPH oxidase, and inflammasome complex activation were all effectively attenuated by Tpl2 inhibition. Tpl2 inhibition by NA also effectively reduced inflammatory/angiogenic factors, retinal leukostasis in streptozotocin-induced diabetic rats, and RPE secretion of inflammatory cytokines. The attenuated release of angiogenic factors led to inhibited vascular abnormalities in the diabetic animal model. CONCLUSIONS: The inhibition of Tpl2 can block the inflammasome signaling pathway in RPE and has potential clinical and therapeutic implications in diabetes-associated retinal microvascular dysfunction.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Diabetic Retinopathy/prevention & control , Inflammasomes/antagonists & inhibitors , MAP Kinase Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Retinal Neovascularization/prevention & control , Retinal Pigment Epithelium/drug effects , Aged , Animals , Cells, Cultured , Cross-Sectional Studies , Databases, Factual , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/enzymology , Diabetic Retinopathy/enzymology , Diabetic Retinopathy/etiology , Diabetic Retinopathy/pathology , Female , Humans , Inflammasomes/metabolism , MAP Kinase Kinase Kinases/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Pregnancy , Prospective Studies , Proto-Oncogene Proteins/metabolism , Retinal Neovascularization/enzymology , Retinal Neovascularization/etiology , Retinal Neovascularization/pathology , Retinal Pigment Epithelium/enzymology , Retinal Pigment Epithelium/pathology , Signal Transduction
5.
Circ Res ; 121(6): e37-e52, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28724746

ABSTRACT

RATIONALE: Diabetic retinopathy is characterized by vasopermeability, vascular leakage, inflammation, blood-retinal barrier breakdown, capillary degeneration, and neovascularization. However, the mechanisms underlying the association between diabetes mellitus and progression retinopathy remain unclear. OBJECTIVE: TPL2 (tumor progression locus 2), a serine-threonine protein kinase, exerts a pathological effect on vascular angiogenesis. This study investigated the role of Nε-(carboxymethyl)lysine, a major advanced glycation end products, and the involved TPL2-related molecular signals in diabetic retinopathy using models of in vitro and in vivo and human samples. METHODS AND RESULTS: Serum Nε-(carboxymethyl)lysine levels and TPL2 kinase activity were significantly increased in clinical patients and experimental animals with diabetic retinopathy. Intravitreal administration of pharmacological blocker or neutralizing antibody inhibited TPL2 and effectively suppressed the pathological characteristics of retinopathy in streptozotocin-induced diabetic animal models. Intravitreal VEGF (vascular endothelial growth factor) neutralization also suppressed the diabetic retinopathy in diabetic animal models. Mechanistic studies in primary human umbilical vein endothelial cells and primary retinal microvascular endothelial cells from streptozotocin-diabetic rats, db/db mice, and samples from patients with diabetic retinopathy revealed a positive parallel correlation between Nε-(carboxymethyl)lysine and the TPL2/chemokine SDF1α (stromal cell-derived factor-α) axis that is dependent on endoplasmic reticulum stress-related molecules, especially ATF4 (activating transcription factor-4). CONCLUSIONS: This study demonstrates that inhibiting the Nε-(carboxymethyl)lysine-induced TPL2/ATF4/SDF1α axis can effectively prevent diabetes mellitus-mediated retinal microvascular dysfunction. This signaling axis may include the therapeutic potential for other diseases involving pathological neovascularization or macular edema.


Subject(s)
Activating Transcription Factor 4/metabolism , Chemokine CXCL12/metabolism , Diabetic Retinopathy/metabolism , MAP Kinase Kinase Kinases/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lysine/analogs & derivatives , Lysine/blood , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Signal Transduction
6.
Oncotarget ; 8(10): 16925-16938, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-28129651

ABSTRACT

Excessive deposition of extracellular matrix (ECM) in the glomerulus contributed by mesangial cells is the hallmark of diabetic nephropathy, eventually leading to glomerulosclerosis. In this study, we examined the regulatory signals involved in the high glucose (HG)-induced overproduction of ECM in rat mesangial cells (RMCs). We disclosed excessive fibronectin and collagen IV production, tyrosine phosphorylation of signal transducer and activator of transcription 1 and 3 (STAT1/3), and up-regulation of suppressor of cytokine signaling-3 (SOCS-3) expression in HG-treated RMCs. STAT1/STAT3 binding element was essential for SOCS-3 promoter activity stimulated by HG. HG was capable of promoting the specific DNA binding activities to an oligonucleotide probe containing the SOCS-3 sequence. The selective phosphoinositide 3-kinase (PI3K) inhibitor LY294002 and dominant negative p85 vector (DNΔp85) transfection effectively abolished these HG-induced responses. Moreover, HG markedly increased the cyclin kinase inhibitor p27Kip1 protein expression, which could be inhibited by LY294002 or transfection of DNΔp85. Taken together, these results suggest that HG-induced SOCS-3 upregulation depends upon the presence of STAT-binding element in the SOCS-3 promoter, which is specifically activated by STAT1/3. The PI3K/STAT1/3 signaling pathway mediated HG-triggered ECM accumulation and SOCS-3 upregulation in RMCs.


Subject(s)
Extracellular Matrix/drug effects , Glucose/pharmacology , Mesangial Cells/drug effects , Phosphatidylinositol 3-Kinases/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Animals , Base Sequence , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Extracellular Matrix/metabolism , Immunoblotting , Male , Mesangial Cells/metabolism , Phosphatidylinositol 3-Kinases/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Rats, Sprague-Dawley , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein/genetics , Time Factors
7.
Oncotarget ; 8(12): 19376-19388, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28038445

ABSTRACT

Ochratoxin A (OTA) contaminated food increases reactive oxygen species (ROS) production in glomerulus and causes glomerulopathy. The molecular mechanisms still remain uncertain. In this study, we used mouse and rat glomerular mesangial cells and delineate the signaling pathway behind the OTA-triggered cell apoptosis. OTA dose-dependently induced expression of ER stress markers including phospho-PERK, phospho-eIF2α, GRP78, GRP94, and CHOP. Apoptosis events including cleavage of caspase-12, caspase-7, and PARP are also observed. OTA activated oxidative stress and increased NADPH oxidase activity. NADPH oxidase inhibitor, apocynin, significantly attenuated OTA-induced cell apoptosis. Moreover, OTA markedly increased the calpain activity which significantly inhibited by apocynin. Transfection of calpain-siRNA effectively inhibited the OTA-increased ER stress-related protein expression. These findings suggest that OTA activated NADPH oxidase and calpain, induced ER stress and ROS production, and caused glomerular mesangial cells apoptosis which leads to glomerulopathy.


Subject(s)
Apoptosis/drug effects , Calpain/metabolism , Endoplasmic Reticulum Stress/drug effects , Mesangial Cells/pathology , NADPH Oxidases/metabolism , Ochratoxins/pharmacology , Reactive Oxygen Species/metabolism , Animals , Calcium Channel Blockers/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Signal Transduction
8.
Chemistry ; 22(30): 10620-6, 2016 Jul 18.
Article in English | MEDLINE | ID: mdl-27346677

ABSTRACT

Herein, mesoporous sodium vanadium phosphate nanoparticles with highly sp(2) -coordinated carbon coatings (meso-Na3 V2 (PO4 )3 /C) were successfully synthesized as efficient cathode material for rechargeable sodium-ion batteries by using ascorbic acid as both the reductant and carbon source, followed by calcination at 750 °C in an argon atmosphere. Their crystalline structure, morphology, surface area, chemical composition, carbon nature and amount were systematically explored. Following electrochemical measurements, the resultant meso-Na3 V2 (PO4 )3 /C not only delivered good reversible capacity (98 mAh g(-1) at 0.1 A g(-1) ) and superior rate capability (63 mAh g(-1) at 1 A g(-1) ) but also exhibited comparable cycling performance (capacity retention: ≈74 % at 450 cycles at 0.4 A g(-1) ). Moreover, the symmetrical sodium-ion full cell with excellent reversibility and cycling stability was also achieved (capacity retention: 92.2 % at 0.1 A g(-1) with 99.5 % coulombic efficiency after 100 cycles). These attributes are ascribed to the distinctive mesostructure for facile sodium-ion insertion/extraction and their continuous sp(2) -coordinated carbon coatings, which facilitate electronic conduction.

9.
J Pineal Res ; 60(2): 142-54, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26514342

ABSTRACT

Peritoneal dissemination of tumor has high mortality and is associated with the loss of epithelial features, acquisition of motile mesenchymal morphology characteristics, and invasive properties by tumor cells. Melatonin is an endogenously produced molecule in all plant species that is known to exert antitumor activity, but to date, its underlying mechanisms and antiperitoneal metastasis efficacy is not well defined. This study determined the antiperitoneal dissemination potential of melatonin in vivo and assessed its association with the inhibition of epithelial-to-mesenchymal transition (EMT) signaling mechanism by endoplasmic reticulum (ER) stress, which may be a major molecular mechanism of melatonin against cancer. The results demonstrate that melatonin inhibited peritoneal metastasis in vivo and activated ER stress in Cignal ERSE Reporter Assay, organelle structure in transmission electron microscopy images, calpain activity, and protein biomarkers like p-elf2α. Moreover, the overexpression of transcription factor C/EBPß in gastric cancer interacted with NFκB and further regulates COX-2 expression. These were dissociated and downregulated by melatonin, as proven by immunofluorescence imaging, immunoprecipitation, EMSA, and ChIP assay. Melatonin or gene silencing of C/EBPß decreased the EMT protein markers (E-cadherin, Snail, and Slug) and Wnt/beta-catenin activity by Topflash activity, and increased ER stress markers. In an animal study, the results of melatonin therapy were consistent with those of in vitro findings and attenuated systemic proangiogenesis factor production. In conclusion, C/EBPß and NFκB inhibition by melatonin may impede both gastric tumor growth and peritoneal dissemination by inducing ER stress and inhibiting EMT.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Calpain/metabolism , Endoplasmic Reticulum Stress/drug effects , Epithelial-Mesenchymal Transition/drug effects , Melatonin/pharmacology , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Peritoneal Neoplasms/drug therapy , Proteolysis/drug effects , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Calpain/genetics , Cell Line, Tumor , Gene Silencing , Humans , Mice , NF-kappa B/genetics , Neoplasm Proteins/genetics , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
10.
Oncotarget ; 5(17): 7788-804, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25226618

ABSTRACT

Biseugenol (Eug) is known to antiproliferative of cancer cells; however, to date, the antiperitoneal dissemination effects have not been studied in any mouse cancer model. In this study, Aryl hydrocarbon receptor (AhR) expression was associated with lymph node and distant metastasis in patients with gastric cancer and was correlated with clinicolpathological pattern. We evaluated the antiperitoneal dissemination potential of knockdown AhR and Biseugenol in cancer mouse model and assessed mesenchymal characteristics. Our results demonstrate that tumor growth, peritoneal dissemination and peritoneum or organ metastasis implanted MKN45 cells were significantly decreased in shAhR and Biseugenol-treated mice and that endoplasmic reticulum (ER) stress was caused. Biseugenol-exposure tumors showed acquired epithelial features such as phosphorylation of E-cadherin, cytokeratin-18 and loss mesenchymal signature Snail, but not vimentin regulation. Snail expression, through AhR activation, is an epithelial-to-mesenchymal transition (EMT) determinant. Moreover, Biseugenol enhanced Calpain-10 (Calp-10) and AhR interaction results in Snail downregulation. The effect of shCalpain-10 in cancer cells was associated with inactivation of AhR/Snail promoter binding activity. Inhibition of Calpain-10 in gastric cancer cells by short hairpin RNA or pharmacological inhibitor was found to effectively reduced growth ability and vessel density in vivo. Importantly, knockdown of AhR completed abrogated peritoneal dissemination. Herein, Biseugenol targeting ER stress provokes Calpain-10 activity, sequentially induces reversal of EMT and apoptosis via AhR may involve the paralleling processes. Taken together, these data suggest that Calpain-10 activation and AhR inhibition by Biseugenol impedes both gastric tumor growth and peritoneal dissemination by inducing ER stress and inhibiting EMT.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Peritoneal Neoplasms/secondary , Receptors, Aryl Hydrocarbon/biosynthesis , Stomach Neoplasms/pathology , Aged , Animals , Chromatin Immunoprecipitation , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Endoplasmic Reticulum Stress/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Male , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Middle Aged , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Xenograft Model Antitumor Assays
11.
Free Radic Biol Med ; 74: 294-306, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25014566

ABSTRACT

N(ε)-carboxymethyllysine (CML) is an important driver of diabetic vascular complications and endothelial cell dysfunction. However, how CML dictates specific cellular responses and the roles of protein tyrosine phosphatases and ERK phosphorylation remain unclear. We examined whether endoplasmic reticulum (ER) localization of MAPK phosphatase-3 (MKP-3) is critical in regulating ERK inactivation and promoting NADPH oxidase-4 (Nox4) activation in CML-induced endothelial cell injury. We demonstrated that serum CML levels were significantly increased in type 2 diabetes patients and diabetic animals. CML induced ER stress and apoptosis, reduced ERK activation, and increased MKP-3 protein activity in HUVECs and SVECs. MKP-3 siRNA transfection, but not that of MKP-1 or MKP-2, abolished the effects of CML on HUVECs. Nox4-mediated activation of MKP-3 regulated the switch to ERK dephosphorylation. CML also increased the integration of MKP-3 with ERK, which was blocked by silencing MKP-3. Exposure of antioxidants abolished CML-increased MKP-3 activity and protein expression. Furthermore, immunohistochemical staining of both MKP-3 and CML was increased, but phospho-ERK staining was decreased in the aortic endothelium of streptozotocin-induced and high-fat diet-induced diabetic mice. Our results indicate that an MKP-3 pathway might regulate ERK dephosphorylation through Nox4 during CML-triggered endothelial cell dysfunction/injury, suggesting that therapeutic strategies targeting the Nox4/MKP-3 interaction or MKP-3 activation may have clinical implications for diabetic vascular complications.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Dual Specificity Phosphatase 6/metabolism , Endoplasmic Reticulum Stress , Endothelial Cells/drug effects , Lysine/analogs & derivatives , NADPH Oxidases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Transformed , Diabetes Mellitus, Type 2/chemically induced , Diet, High-Fat , Dual Specificity Phosphatase 6/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Endothelial Cells/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Lysine/blood , Lysine/pharmacology , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , NADPH Oxidase 4 , Protein Binding/drug effects , Protein Binding/genetics , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
12.
Nanoscale ; 5(24): 12115-9, 2013 Dec 21.
Article in English | MEDLINE | ID: mdl-24150659

ABSTRACT

This study reports the successful synthesis of ternary spinel-based ZnCo2O4 nanoflakes (NFs) with mesoporous architectures via the combination of a urea-assisted hydrothermal reaction with calcination in an air atmosphere. Owing to their favorable mesostructures and desirable bifunctional oxygen reduction and evolution activities, the resulting mesoporous ZnCo2O4 NFs yielded stable cyclability at a cut-off capacity of 500 mA h gcarbon(-1) in the case of aprotic Li-O2 batteries.

13.
Chemistry ; 19(45): 15358-65, 2013 Nov 04.
Article in English | MEDLINE | ID: mdl-24115190

ABSTRACT

Novel structures of luminescent materials, which are used as light sources for next-generation illumination, are continuously being improved for use in white-light-emitting diodes. Activator-doped known structures are reported as habitual down-conversion phosphors in solid-state lightings and displays. Consequently, the intrinsic qualities of the existent compounds produce deficiencies that limit their applications. Herein we report a spiral-network single-crystal orthophosphate (LiSrPO4) prepared in a platinum crucible with LiCl flux through crystal-growth reactions of SrCl2 and Li3PO4 in air. It crystallizes in a hexagonal system with a=5.0040(2) and c=24.6320(16) Å, V=534.15(5) Å(3), and Z=6 in the space group P6(5). The unit cell is comprised of LiO4 and PO4 tetrahedrons that form a three-dimensional LiPO4(2-) anionic framework with a helical channel structure along the c axis in which the Sr(2+) cation is accommodated. The optical band gap of this composition is about 3.65 eV, as determined by using UV/Vis absorption and diffuse reflection spectra. We used the crystal-growth method to synthesize blue- and red-emitting crystals that exhibited pure color, low reabsorption, a large Stokes shift, and efficient conversion of ultraviolet excitation light into visible light. Emphasis was placed on the development of gratifying structure-related properties of rare-earth luminescent materials and their applications.

14.
Eur J Immunol ; 43(11): 2854-65, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23881867

ABSTRACT

In response to aggravation by activated microglia, IL-13 can significantly enhance ER stress induction, apoptosis, and death via reciprocal signaling through CCAAT/enhancer-binding protein alpha (C/EBP-α) and C/EBP-beta (C/EBP-ß). This reciprocal signaling promotes neuronal survival. Since the induction of cyclooxygenase-2 (COX-2) and peroxisome proliferator-activated receptor gamma/heme oxygenase 1 (PPAR-γ/HO-1) by IL-13 plays a crucial role in the promotion of and protection from activated microglia, respectively; here, we investigated the role of IL-13 in regulating C/EBPs in activated microglia and determined its correlation with neuronal function. The results revealed that IL-13 significantly enhanced C/EBP-α/COX-2 expression and PGE2 production in LPS-treated microglial cells. Paradoxically, IL-13 abolished C/EBP-ß/PPAR-γ/HO-1 expression. IL-13 also enhanced ER stress-evoked calpain activation by promoting the association of C/EBP-ß and PPAR-γ. SiRNA-C/EBP-α effectively reversed the combined LPS-activated caspase-12 activation and IL-13-induced apoptosis. In contrast, siRNA-C/EBP-ß partially increased microglial cell apoptosis. By NeuN immunochemistry and CD11b staining, there was improvement in the loss of CA3 neuronal cells after intrahippocampal injection of IL-13. This suggests that IL-13-enhanced PLA2 activity regulates COX-2/PGE2 expression through C/EBP-α activation. In parallel, ER stress-related calpain downregulates the PPAR-γ/HO-1 pathway via C/EBP-ß and leads to aggravated death of activated microglia via IL-13, thereby preventing cerebral inflammation and neuronal injury.


Subject(s)
Apoptosis , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Interleukin-13/metabolism , Microglia/metabolism , Neurons/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/genetics , Calpain/metabolism , Caspase 12/metabolism , Cells, Cultured , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Heme Oxygenase-1/biosynthesis , Lipopolysaccharides , Membrane Proteins/biosynthesis , PPAR gamma/metabolism , RNA Interference , RNA, Small Interfering , Rats , Signal Transduction
15.
Carcinogenesis ; 34(11): 2568-79, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23828905

ABSTRACT

Honokiol is known to suppress the growth of cancer cells; however, to date, its antiperitoneal dissemination effects have not been studied in an orthotopic mouse model. In the present study, we evaluated the antiperitoneal dissemination potential of Honokiol in an orthotopic mouse model and assessed associations with tumor growth factor-ß1 (TGFß1) and cells stimulated by a carcinogen, N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Our results demonstrate that tumor growth, peritoneal dissemination and peritoneum or organ metastasis of orthotopically implanted MKN45 cells were significantly decreased in Honokiol-treated mice and that endoplasmic reticulum (ER) stress was induced. Honokiol-treated tumors showed increased epithelial signatures such as E-cadherin, cytokeratin-18 and ER stress marker. In contrast, decreased expression of vimentin, Snail and tumor progression locus 2 (Tpl2) was also noted. TGFß1 and MNNG-induced downregulation of E-cadherin and upregulation of Tpl2 were abrogated by Honokiol treatment. The effect of Tpl2 inhibition in cancer cells or endothelial cells was associated with inactivation of CCAAT/enhancer binding protein B, nuclear factor kappa-light-chain-enhancer of activated B cell and activator protein-1 and suppression of vascular endothelial growth factor. Inhibition of Tpl2 in gastric cancer cells by small interfering RNA or pharmacological inhibitor was found to effectively reduce growth ability and vessel density in vivo. Honokiol-induced reversal of epithelial-to-mesenchymal transition (EMT) and ER stress-induced apoptosis via Tp12 may involve the paralleling processes. Taken together, our results suggest that the therapeutic inhibition of Tpl2 by Honokiol thwarts both gastric tumor growth and peritoneal dissemination by inducing ER stress and inhibiting EMT.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biphenyl Compounds/pharmacology , Lignans/pharmacology , MAP Kinase Kinase Kinases/metabolism , Peritoneal Neoplasms/prevention & control , Phytotherapy , Proto-Oncogene Proteins/metabolism , Stomach Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Luciferases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured
16.
J Pathol ; 230(2): 215-27, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22553146

ABSTRACT

N(ε)-carboxymethyllysine (CML), a major advanced glycation end product, plays a crucial role in diabetes-induced vascular injury. The roles of protein tyrosine phosphatases and vascular endothelial growth factor (VEGF) receptors in CML-related endothelial cell injury are still unclear. Human umbilical vein endothelial cells (HUVECs) are a commonly used human EC type. Here, we tested the hypothesis that NADPH oxidase/reactive oxygen species (ROS)-mediated SH2 domain-containing tyrosine phosphatase-1 (SHP-1) activation by CML inhibits the VEGF receptor-2 (VEGFR-2, KDR/Flk-1) activation, resulting in HUVEC injury. CML significantly inhibited cell proliferation and induced apoptosis and reduced VEGFR-2 activation in parallel with the increased SHP-1 protein expression and activity in HUVECs. Adding recombinant VEGF increased forward biological effects, which were attenuated by CML. The effects of CML on HUVECs were abolished by SHP-1 siRNA transfection. Exposure of HUVECs to CML also remarkably escalated the integration of SHP-1 with VEGFR-2. Consistently, SHP-1 siRNA transfection and pharmacological inhibitors could block this interaction and elevating [(3)H]thymidine incorporation. CML also markedly activated the NADPH oxidase and ROS production. The CML-increased SHP-1 activity in HUVECs was effectively attenuated by antioxidants. Moreover, the immunohistochemical staining of SHP-1 and CML was increased, but phospho-VEGFR-2 staining was decreased in the aortic endothelium of streptozotocin-induced and high-fat diet-induced diabetic mice. We conclude that a pathway of tyrosine phosphatase SHP-1-regulated VEGFR-2 dephosphorylation through NADPH oxidase-derived ROS is involved in the CML-triggered endothelial cell dysfunction/injury. These findings suggest new insights into the development of therapeutic approaches to reduce diabetic vascular complications.


Subject(s)
Endothelium, Vascular/drug effects , Glycation End Products, Advanced/pharmacology , Lysine/analogs & derivatives , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Diabetes Mellitus, Experimental/metabolism , Endothelium, Vascular/metabolism , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Lysine/pharmacology , Male , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Transfection
17.
PLoS One ; 7(8): e43711, 2012.
Article in English | MEDLINE | ID: mdl-22937084

ABSTRACT

BACKGROUND: Honokiol, a small-molecular weight natural product, has previously been reported to activate apoptosis and inhibit gastric tumorigenesis. Whether honokiol inhibits the angiogenesis and metastasis of gastric cancer cells remains unknown. METHODOLOGY/PRINCIPAL FINDINGS: We tested the effects of honokiol on angiogenic activity and peritoneal dissemination using in vivo, ex vivo and in vitro assay systems. The signaling responses in human gastric cancer cells, human umbilical vascular endothelial cells (HUVECs), and isolated tumors were detected and analyzed. In a xenograft gastric tumor mouse model, honokiol significantly inhibited the peritoneal dissemination detected by PET/CT technique. Honokiol also effectively attenuated the angiogenesis detected by chick chorioallantoic membrane assay, mouse matrigel plug assay, rat aortic ring endothelial cell sprouting assay, and endothelial cell tube formation assay. Furthermore, honokiol effectively enhanced signal transducer and activator of transcription (STAT-3) dephosphorylation and inhibited STAT-3 DNA binding activity in human gastric cancer cells and HUVECs, which was correlated with the up-regulation of the activity and protein expression of Src homology 2 (SH2)-containing tyrosine phosphatase-1 (SHP-1). Calpain-II inhibitor and siRNA transfection significantly reversed the honokiol-induced SHP-1 activity. The decreased STAT-3 phosphorylation and increased SHP-1 expression were also shown in isolated peritoneal metastatic tumors. Honokiol was also capable of inhibiting VEGF generation, which could be reversed by SHP-1 siRNA transfection. CONCLUSIONS/SIGNIFICANCE: Honokiol increases expression and activity of SPH-1 that further deactivates STAT3 pathway. These findings also suggest that honokiol is a novel and potent inhibitor of angiogenesis and peritoneal dissemination of gastric cancer cells, providing support for the application potential of honokiol in gastric cancer therapy.


Subject(s)
Biphenyl Compounds/pharmacology , Calpain/metabolism , Lignans/pharmacology , Neovascularization, Pathologic/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/pathology , Animals , Apoptosis/drug effects , Calpain/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Mice , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RNA, Small Interfering , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
18.
Dalton Trans ; 41(38): 11885-8, 2012 Oct 14.
Article in English | MEDLINE | ID: mdl-22914856

ABSTRACT

A polymorph of a [Zn(2)(IM)(4)·(DMF)] (IM: imidazolate; DMF: dimethyl formamide) framework was synthesized using a DMF template. The topology of a ZnN(4) framework is similar to that of zeolites with a chain of connected SiO(4) tetrahedra. This polymorph has a zeolite-like structure and a wave shape topology in the [010] direction. The main chain is connected to an imidazolate ligand. The structure has excellent chemical and thermal stability. This framework also exhibits broad range near-UV excitation from 350 nm to 430 nm and broadened photoluminescence emission at 445 nm. It has great potential as a phosphor in applications of near-UV or UV white light-emitting diodes.

19.
Br J Pharmacol ; 160(8): 1963-72, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20649594

ABSTRACT

BACKGROUND AND PURPOSE: Peroxisome proliferator-activated receptor-gamma (PPAR-gamma), COX-2 and 15-lipoxygenase (LOX)-1 have been shown to be involved in tumour growth. However, the roles of PPAR-gamma, COX-2 or 15-LOX-1 in gastric tumourigenesis remain unclear. Here, we investigate the role of 15-LOX-1 induction by honokiol, a small-molecular weight natural product, in PPAR-gamma and COX-2 signalling during gastric tumourigenesis. EXPERIMENTAL APPROACH: Human gastric cancer cell lines (AGS, MKN45, N87 and SCM-1) were cultured with or without honokiol. Gene and protein expressions were analysed by RT-PCR and Western blotting respectively. Small interfering RNAs (siRNAs) for COX-2, PPAR-gamma and 15-LOX-1 were used to interfere with the expressions of these genes. A xenograft gastric tumour model in mouse was used for in vivo study. KEY RESULTS: PPAR-gamma and COX-2 proteins were highly expressed in gastric cancer cells. Inhibitors, or siRNA for COX-2 or PPAR-gamma, significantly decreased cell viability. Honokiol markedly inhibited PPAR-gamma and COX-2 expressions in gastric cancer cells and tumours of xenograft mice, and induced apoptosis and cell death. Honokiol markedly activated cellular 15-LOX-1 expression and 13-S-hydroxyoctadecadienoic acid (a primary product of 15-LOX-1 metabolism of linoleic acid) production. 15-LOX-1 siRNA could reverse the honokiol-induced down-regulation of PPAR-gamma and COX-2, and cell apoptosis. 15-LOX-1 was markedly induced in tumours of xenograft mice treated with honokiol. CONCLUSIONS AND IMPLICATIONS: These findings suggest that induction of 15-LOX-1-mediated down-regulation of a PPAR-gamma and COX-2 pathway by honokiol may be a promising therapeutic strategy for gastric cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Arachidonate 15-Lipoxygenase/metabolism , Biphenyl Compounds/pharmacology , Cyclooxygenase 2/metabolism , Lignans/pharmacology , PPAR gamma/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Arachidonate 15-Lipoxygenase/genetics , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Linoleic Acids/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , PPAR gamma/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...