Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 8(53): 91258-91269, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29207641

ABSTRACT

Here we report that a novel synthesized compound (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) which exhibits better stability, drug-likeness and anti-cancer effect than (E)-2,4-bis(p-hydroxyphenyl)-2-butenal (BHPB) that we previously reported. Of all newly synthesized BHPB analogues, MMPP showed the most significant inhibitory effect on colon cancer cell growth. Thus, we evaluated the anti-cancer effects and possible mechanisms of MMPP in vitro and in vivo. MMPP treatment (0-15 µg/mL) induced apoptotic cell death and enhanced the expression of cleaved caspase-3 and cleaved caspase-8 in a concentration dependent manner. Notably, the expression of death receptor (DR)5 and DR6 was significantly increased by MMPP treatment. Moreover, DR5 siRNA or DR6 siRNA transfection partially abolished MMPP-induced cell growth inhibition. Pull down assay and docking experiment showed that MMPP bound directly to IkappaB kinase ß (IKKß). It was noteworthy that IKKß mutant (C99S) partially abolished MMPP-induced cell growth inhibition and enhanced expression of DR5 and DR6. In addition, MMPP enhanced TRAIL-induced apoptosis, cell growth inhibition and expression of DRs. In xenograft mice model, MMPP (2.5-5 mg/kg) suppressed tumor growth in a dose dependent manner. Immunohistochemistry analysis showed that the expression levels of DR5 and DR6 and active caspase-3 were increased while the expression levels of PCNA and p-IKKß were decreased in a dose dependent manner. Thus, MMPP may be a promising anti-cancer agent in colon cancer treatment.

2.
Oncotarget ; 7(11): 11984-99, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26910914

ABSTRACT

Chemokine receptors are implicated in inflammation and immune responses. Neuro-inflammation is associated with activation of astrocyte and amyloid-beta (Aß) generations that lead to pathogenesis of Alzheimer disease (AD). Previous our study showed that deficiency of CC chemokine receptor 5 (CCR5) results in activation of astrocytes and Aß deposit, and thus memory dysfunction through increase of CC chemokine receptor 2 (CCR2) expression. CCR5 knockout mice were used as an animal model with memory dysfunction. For the purpose LPS was injected i.p. daily (0.25 mg/kg/day). The memory dysfunctions were much higher in LPS-injected CCR5 knockout mice compared to CCR5 wild type mice as well as non-injected CCR5 knockout mice. Associated with severe memory dysfuction in LPS injected CCR5 knockout mice, LPS injection significant increase expression of inflammatory proteins, astrocyte activation, expressions of ß-secretase as well as Aß deposition in the brain of CCR5 knockout mice as compared with that of CCR5 wild type mice. In CCR5 knockout mice, CCR2 expressions were high and co-localized with GFAP which was significantly elevated by LPS. Expression of monocyte chemoattractant protein-1 (MCP-1) which ligands of CCR2 also increased by LPS injection, and increment of MCP-1 expression is much higher in CCR5 knockout mice. BV-2 cells treated with CCR5 antagonist, D-ala-peptide T-amide (DAPTA) and cultured astrocytes isolated from CCR5 knockout mice treated with LPS (1 µg/ml) and CCR2 antagonist, decreased the NF-ĸB activation and Aß level. These findings suggest that the deficiency of CCR5 enhances response of LPS, which accelerates to neuro-inflammation and memory impairment.


Subject(s)
Astrocytes/pathology , Gliosis/etiology , Inflammation/complications , Lipopolysaccharides/toxicity , Memory Disorders/etiology , Plaque, Amyloid/etiology , Receptors, CCR5/physiology , Animals , Apoptosis , Astrocytes/drug effects , Behavior, Animal , Cell Proliferation , Cells, Cultured , Gliosis/pathology , Inflammation/chemically induced , Male , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Amyloid/pathology
3.
Mol Carcinog ; 55(5): 659-70, 2016 May.
Article in English | MEDLINE | ID: mdl-25865242

ABSTRACT

Naphthofuran compounds have been known to regulate HNF 4α which is associated with proliferation, progression and metastasis of HCC. In this study, we investigated whether N-(3,5-bis(trifluoromethyl)phenyl)-5-chloro-2,3-dihydronaphtho[1,2-b]furan-2-carboxamide (NHDC), a novel synthetic naphthofuran compound inhibits liver tumor growth through activation of HNF 4α. Treatment with different concentrations (1-10.8 µM) of NHDC for various periods (0-72 h) inhibited liver cancer cells (HepG2, Hep3B) growth as well as colony formation followed by induction of apoptosis in a concentration dependent manner. NHDC also induced expression of the apoptosis regulating genes as well as inhibiting the action of STAT3. These inhibitory effects were associated with enhancement of expression and DNA binding activity of HNF 4α. In vivo study confirmed that liver tumor growth was prevented with NHDC (5 mg/kg), and its effect was also related with inhibition of STAT3 pathway through enhancement of expression and DNA binding activity of HNF 4α. Moreover, siRNA of HNF 4α abolished NHDC-induced cell growth inhibition as well as DNA binding activity and phosphorylation of STAT3. Pull down assay docking prediction analysis proved that NHDC directly binds to hydrophobic fatty acid ligand binding site of HNF 4α. A novel naphthofuran compound, NHDC inhibited liver tumor growth by inactivating of STAT3 through direct biding to HNF 4α.


Subject(s)
Antineoplastic Agents/administration & dosage , Furans/administration & dosage , Hepatocyte Nuclear Factor 4/metabolism , Liver Neoplasms/drug therapy , Naphthalenes/administration & dosage , Naphthols/administration & dosage , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Furans/chemical synthesis , Furans/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Mice , Naphthalenes/chemical synthesis , Naphthalenes/pharmacology , Naphthols/chemical synthesis , Naphthols/pharmacology , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
4.
Mol Neurobiol ; 52(1): 176-86, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25128029

ABSTRACT

Estrogen has been known to reduce the development of Alzheimer's disease (AD). However, exact mechanisms are not clear. We investigated whether estrogen can increase amyloid-beta (Aß) degradation and affects Aß-induced memory impairment in an estrogen deficiency model. Estrogen receptor alpha (ERα) knockout mice and wild-type mice were intracerebroventricular (ICV) infused with Aß (300 pmol) for 2 weeks. Cognitive function was then assessed by the Morris water maze test and passive avoidance test. In addition, Western blot analysis, immunostaining, immunofluorescence staining, ELISA, and enzyme activity assays were used to examine the degree of Aß deposition in the brains of ERα knockout mice. In our present study, Aß was accumulated more in the ERα knockout mice brain and greatly worsened memory impairment and glial activation as well as neurogenic inflammation. These results suggest that estrogen may protect memory impairment by stimulating the degradation of Aß and down-regulate neurogenic inflammation as well as amyloidogenesis.


Subject(s)
Amyloid beta-Peptides/metabolism , Estrogen Receptor alpha/deficiency , Memory Disorders/metabolism , Amyloid/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Death , Cyclooxygenase 2/metabolism , Estrogen Receptor alpha/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Liver/metabolism , Liver/pathology , Low Density Lipoprotein Receptor-Related Protein-1 , Matrix Metalloproteinase 9/metabolism , Memory Disorders/pathology , Mice, Inbred C57BL , Mice, Knockout , Neprilysin/metabolism , Nitric Oxide Synthase Type II/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism
5.
Mol Neurobiol ; 51(2): 648-60, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24854197

ABSTRACT

Neuroinflammation is important for the development of several neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and stroke. Since changes of cytokine level are critical for neuroinflammation in the brain, we investigated whether IL-32α overexpression could change neuroinflammation and, thus, affect stroke development. Middle cerebral artery occlusion (MCAO) induced development of ischemia, and ischemic neuronal cell death were reduced in IL-32α-overexpressing transgenic mice (IL-32α mice) brain through the decreased release of neuroinflammatory cytokines (IL-6, IL-1ß, TNF-α) and activation of astrocytes, but enhancement of anti-neuroinflammatory cytokines (IL-10). Reactive oxygen species generation and lipid peroxidation as well as expression of inducible nitric oxide and cyclooxygenase-2 were also reduced in the IL-32α mice brain. Nuclear factor-kappa B (NF-κB), a critical transcriptional factor regulating neuroinflammation, was much lower, but activation of signal transducer and activator of transcription 3 (STAT3), which plays a crucial role in cell survival and proliferation, was much higher in IL-32α-overexpressing mice brain compared to those of wild-type mice brain. These results suggest that IL-32α can prevent cerebral ischemia damage via upregulation of anti-neuroinflammatory cytokine expression and STAT3 activation, but downregulation of neuroinflammatory cytokines and NF-κB activation.


Subject(s)
Interleukins/biosynthesis , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , STAT3 Transcription Factor/biosynthesis , Signal Transduction/physiology , Stroke/metabolism , Stroke/prevention & control , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
J Neuroinflammation ; 11: 118, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24985096

ABSTRACT

BACKGROUND: ent-Sauchinone is a polyphenolic compound found in plants belonging to the lignan family. ent-Sauchinone has been shown to modulate the expression of inflammatory factors through the nuclear factor-kappa B (NF-κB) signaling pathway. It is well known that neuroinflammation is associated with amyloidogenesis. Thus, in the present study, we investigated whether ent-Sauchinone could have anti-amyloidogenic effects through the inhibition of NF-κB pathways via its anti-inflammatory property. METHODS: To investigate the potential effect of ent-Sauchinone on anti-neuroinflammation and anti-amyloidogenesis in in vitro studies, we used microglial BV-2 cells and cultured astrocytes treated with ent-Sauchinone (1, 5, and 10 µM) for 24 hours. For the detection of anti-neuro-inflammatory responses, reative oxygen species (ROS) and Nitric oxide (NO) generation and inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression were measured with assay kits and western blotting. ß-secretase and ß-secretase activities and ß-amyloid levels were determined for measuring the anti-amyloidogenic effects of ent-Sauchinone by enzyme assay kits. NF-κB and STAT3 signals were detected with electromobility shift assay (EMSA) to study the related signaling pathways. The binding of ent-Sauchinone to STAT3 was evaluated by a pull-down assay and by a docking model using Autodock VINA software (Hoover's Inc., Texas, United states). RESULTS: ent-Sauchinone (1, 5, and 10 µM) effectively decreased lipopolysaccharide (LPS)-(1 µg/ml) induced inflammatory responses through the reduction of ROS and NO generations and iNOS and COX-2 expressions in cultured astrocytes and microglial BV-2 cells. ent-Sauchinone also inhibited LPS-induced amyloidogenesis through the inhibition of ß-secretase and ß-secretase activity. NF- κB amyloid and STAT3, critical transcriptional factors regulating not only inflammation but also amyloidogenesis, were also inhibited in a concentration dependent manner by ent-Sauchinone by blocking the phosphorylation of I κB and STAT3 in cultured astrocytes and microglial BV-2 cells. The docking model approach showed that ent-Sauchinone binds to STAT3, and the employment of a STAT3 inhibitor and siRNA reversed ent-Sauchinone-induced inhibition NF-κB activation and Aß generation. CONCLUSIONS: These results indicated that ent-Sauchinone inhibited neuroinflammation and amyloidogenesis through the inhibition of STAT3-mediated NF-κB activity, and thus could be applied in the treatment of neuro-inflammatory diseases, including Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Astrocytes/drug effects , Benzopyrans/pharmacology , Dioxoles/pharmacology , NF-kappa B/metabolism , Peptide Fragments/metabolism , STAT3 Transcription Factor/metabolism , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Cyclooxygenase 2/metabolism , Electrophoretic Mobility Shift Assay , Glial Fibrillary Acidic Protein/metabolism , Lipopolysaccharides/pharmacology , Microglia/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects
7.
Arch Pharm Res ; 27(3): 357-60, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15089044

ABSTRACT

Four crystal forms of ketorolac have been obtained by recrystallization in organic solvents under variable conditions. Different ketorolac polymorphs and pseudopolymorph were characterized by X-ray powder diffraction crystallography (XRD), Differential scanning calorimetry (DSC) and thermogravimetric analysis (TGA). In the dissolution studies in water at 37 +/- 0.5 degrees C, four crystal forms showed different patterns. The solubility of Form I were the highest. The solubility decreased in rank order: Form I > Form II > Form III > Form IV. Form land Form III were shown to have a good physical stability at room temperature for 60 days. However, Form II is converted to Form III and Form IV is converted to Form I after 60 days storage. Therefore, these observations indicate that crystalline polymorphism for ketorolac is readily inter-convertible and the relationship may have to taken into consideration in the formulation of the drug.


Subject(s)
Ketorolac/pharmacokinetics , Chemistry, Pharmaceutical , Crystallization , Ketorolac/chemistry , Solubility , X-Ray Diffraction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...