Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
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
2.
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
3.
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
4.
Antioxid Redox Signal ; 24(4): 217-231, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26415004

ABSTRACT

AIMS: Activation of glomerular mesangial cells (MCs) and functional changes of renal tubular cells are due to metabolic abnormalities, oxidative stress, and matrix accumulation in the diabetic nephropathy (DN). Aryl hydrocarbon receptor (AhR) activation has been implicated in DN. In this study, we investigated the role of AhR in the pathophysiological processes of DN using AhR knockout (AhRKO) and pharmacological inhibitor α-naphthoflavone mouse models. RESULTS: The increased blood glucose, glucose intolerance, MC activation, macrophage infiltration, and extracellular matrix (ECM) accumulation were significantly attenuated in AhRKO mice with diabetic inducer streptozotocin (STZ) treatment. AhR deficiency by genetic knockout or pharmacological inhibition also decreased the induction of cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2), lipid peroxidation, oxidative stress, NADPH oxidase activity, and N-ɛ-carboxymethyllysine (CML, a major advanced glycation end product) in STZ-induced diabetic mice. CML showed remarkably increased AhR/COX-2 DNA-binding activity, protein-DNA interactions, gene regulation, and ECM formation in MCs and renal proximal tubular cells, which could be reversed by siRNA-AhR transfection. CML-increased AhR nuclear translocation and biological activity in MCs and renal proximal tubular cells could also be effectively attenuated by antioxidants. INNOVATION: We elucidate for the first time that AhR plays an important role in MC activation, macrophage infiltration, and ECM accumulation in DN conferred by oxidative stress. CONCLUSIONS: AhR-regulated COX-2/PGE2 expression and ECM deposition through oxidative stress cascade is involved in the CML-triggered MC activation and macrophage infiltration. These findings suggest new insights into the development of therapeutic approaches to reduce diabetic microvascular complications. Antioxid. Redox Signal. 24, 217-231.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/deficiency , Diabetic Nephropathies/metabolism , Extracellular Matrix/pathology , Macrophages/pathology , Mesangial Cells/pathology , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Benzoflavones/pharmacology , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/genetics , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Lipid Peroxidation/drug effects , Macrophages/metabolism , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Oxidative Stress/drug effects , Receptors, Aryl Hydrocarbon/genetics , Streptozocin
5.
Mol Oncol ; 9(4): 834-49, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25619450

ABSTRACT

Peritoneal dissemination is a major clinical obstacle in gastrointestinal cancer therapy, and it accounts for the majority of cancer-related mortality. Calreticulin (CRT) is over-expressed in gastric tumors and has been linked to poor prognosis. In this study, immunohistochemistry studies revealed that the up-regulation of CRT was associated with lymph node and distant metastasis in patients with gastric cancer specimens. CRT was significantly down-regulated in highly metastatic gastric cancer cell lines and metastatic animal by Honokiol-treated. Small RNA interference blocking CRT by siRNA-CRT was translocated to the cells in the early immunogenic response to Honokiol. Honokiol activated endoplasmic reticulum (ER) stress and down-regulated peroxisome proliferator-activated receptor-γ (PPARγ) activity resulting in PPARγ and CRT degradation through calpain-II activity, which could be reversed by siRNA-calpain-II. The Calpain-II/PPARγ/CRT axis and interaction evoked by Honokiol could be blocked by gene silencing or pharmacological agents. Both transforming growth factor (TGF)-ß1 and N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) induced cell migration, invasion and reciprocal down-regulation of epithelial marker E-cadherin, which could be abrogated by siRNA-CRT. Moreover, Honokiol significantly suppressed MNNG-induced gastrointestinal tumor growth and over-expression of CRT in mice. Knockdown CRT in gastric cancer cells was found to effectively reduce growth ability and metastasis in vivo. The present study provides insight into the specific biological behavior of CRT in epithelial-to-mesenchymal transition (EMT) and metastasis. Taken together, our results suggest that the therapeutic inhibition of CRT by Honokiol suppresses both gastric tumor growth and peritoneal dissemination by dictating early translocation of CRT in immunogenic cell death, activating ER stress, and blocking EMT.


Subject(s)
Biphenyl Compounds/pharmacology , Calreticulin/metabolism , Endoplasmic Reticulum Stress/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lignans/pharmacology , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Adult , Animals , Biomarkers, Tumor/metabolism , Calpain/metabolism , Calreticulin/genetics , Cell Death/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Female , Gene Knockdown Techniques , Humans , Macrophages/drug effects , Macrophages/metabolism , Male , Methylnitronitrosoguanidine , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Neoplasm Invasiveness , PPAR gamma/metabolism , Phagocytosis/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Up-Regulation/drug effects
6.
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
7.
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
8.
Neoplasia ; 15(9): 1036-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24027429

ABSTRACT

Angiogenesis is critical in the development of cancer, which involves several angiogenic factors in its peritoneal dissemination. The role of protein tumor progression locus 2 (Tpl2) in angiogenic factor-related endothelial cell angiogenesis is still unclear. To understand the precise mechanism(s) of Tpl2 inhibition in endothelial cells, this study investigated the role of Tpl2 in mediating angiogenic signals using in vitro, in vivo, and ex vivo models. Results showed that inhibition of Tpl2 inhibitor significantly reduced peritoneal dissemination in a mouse model by positron emission tomography/computed tomography imaging. Simultaneously, inhibiting Tpl2 blocked angiogenesis in tumor nodules and prevented angiogenic factor-induced proliferating cell nuclear antigen (PCNA) in endothelial cells. Vascular endothelial growth factor (VEGF) or chemokine (C-X-C motif) ligand 1 (CXCL1) increased Tpl2 kinase activity and phosphorylation in a dose- and time-dependent manner. Furthermore, Tpl2 inhibition or ablation by siRNA prevented the angiogenic signal-induced tube formation in Matrigel plug assay or aortic ring assay. Inhibiting Tpl2 also prevented the angiogenic factor-induced chemotactic motility and migration of endothelial cells. Tpl2 inhibition by CXCL1 or epidermal growth factor in endothelial cells was associated with inactivation of CCAAT/enhancer binding protein ß, nuclear factor κ light-chain enhancer of activated B cells, and activating protein 1 and suppression of VEGF expression. Thus, Tpl2 inhibitors thwart Tpl2-regulated VEGF by inactivating transcription factors involved in angiogenic factor-triggered endothelial cell angiogenesis. These results suggest that the therapeutic inhibition of Tpl2 may extend beyond cancer and include the treatment of other diseases involving pathologic angiogenesis.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Activating Transcription Factor 1/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Movement , Cell Proliferation , Chemokine CXCL1/metabolism , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , RNA Interference , RNA, Small Interfering , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL