Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmacol Res ; 161: 105146, 2020 11.
Article in English | MEDLINE | ID: mdl-32814173

ABSTRACT

Neovascular age-related macular degeneration (nAMD) is a common cause of irreversible vision loss in the elderly. Anti-vascular endothelial growth factor has been effective in treating pathological ocular neovascularization, but it has limitations including the need for repeated intraocular injections for the maintenance of therapeutic effects in most patients and poor or non-response to this agent in some patients. in vitro cellular studies were conducted using retinal pigment epithelial cell lines (ARPE-19 and hTERT-RPE1), human umbilical vein endothelial cells (HUVECs), and human umbilical vein smooth muscle cells (HUVSMCs). in vivo efficacy of ilimaquinone (IQ) was tested in laser-induced choroidal neovascularization mouse and rabbit models. Tissue distribution study was performed in male C57BL6/J mice. IQ, 4,9-friedodrimane-type sesquiterpenoid isolated from the marine sponge, repressed the expression of angiogenic/inflammatory factors and restored the expression of E-cadherin in retinal pigment epithelial cells by inhibiting the Wnt/ß-catenin pathway. In addition, it selectively inhibited proliferation and tube formation of HUVECs by activating the p53 pathway. Topical and intraperitoneal administration of IQ significantly reduced choroidal neovascularization in rabbits and mice with laser-induced choroidal neovascularization. Notably, IQ by the oral route of exposure was highly permeable to the eyes and suppressed abnormal vascular leakage by downregulation of ß-catenin and stabilization of p53 in vivo. Our findings demonstrate that IQ functions through regulation of p53 and Wnt/ß-catenin pathways with conceivable advantages over existing cytokine-targeted anti-angiogenic therapies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Choroidal Neovascularization/prevention & control , Macular Degeneration/prevention & control , Quinones/pharmacology , Retinal Neovascularization/prevention & control , Retinal Vessels/drug effects , Sesquiterpenes/pharmacology , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Cell Line , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Mice, Inbred C57BL , Rabbits , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology
2.
Leuk Res ; 60: 103-108, 2017 09.
Article in English | MEDLINE | ID: mdl-28772205

ABSTRACT

The aberrant activation of Wnt/ß-catenin signaling is involved in the development of multiple myeloma; thus, this signaling pathway is a potential target for the development of therapeutics for this malignancy. Here, we performed cell-based chemical screening and found that CGK012, a pyranocoumarin compound, suppressed the Wnt3a-CM-mediated activation of ß-catenin response transcription. CGK012 induced ß-catenin phosphorylation at Ser33/Ser37/Thr41, leading to proteasomal degradation and reducing the level of intracellular ß-catenin. Furthermore, CGK012 consistently decreased the amount of ß-catenin and repressed the expression of cyclin D1, c-myc, and axin-2 (downstream target genes of ß-catenin) in RPMI-8226 multiple myeloma cells. In addition, CGK012 inhibited the proliferation of RPMI-8226 cells and promoted apoptosis, as indicated by the increase in the population of Annexin V-FITC-stained cells and caspase-3/7 activity. These findings suggest that CGK012 could exert antiproliferative activity against multiple myeloma cells by attenuating the Wnt/ß-catenin pathway; thus, it may have potential as a therapeutic agent for multiple myeloma treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carbamates/pharmacology , Cell Proliferation/drug effects , Coumarins/pharmacology , Multiple Myeloma/pathology , Pyranocoumarins/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Phosphorylation , Pyranocoumarins/therapeutic use
3.
Drug Metab Dispos ; 45(7): 765-769, 2017 07.
Article in English | MEDLINE | ID: mdl-28461575

ABSTRACT

Highly selective cytochrome P450 CYP2J2 (CYP2J2) inhibitors suitable for reaction phenotyping are currently not available. (7S)-(+)-(4-Nitro-phenyl)-acrylic acid, 8,8-dimethyl-2-oxo-6,7-dihydro-2H,8H-pyrano[3,2-g]chromen-7-yl-ester (LKY-047), a decursin derivative, was synthesized, and its inhibitor potencies toward CYP2J2 as well as other cytochrome P450 (P450) enzymes in human liver microsomes (HLM) were evaluated. LKY-047 was demonstrated to be a strong competitive inhibitor of CYP2J2-mediated astemizole O-demethylase and terfenadine hydroxylase activity, with Ki values of 0.96 and 2.61 µM, respectively. It also acted as an uncompetitive inhibitor of CYP2J2-mediated ebastine hydroxylation with a Ki value of 3.61 µM. Preincubation of LKY-047 with HLMs and NADPH did not alter inhibition potency, indicating that it is not a mechanism-based inhibitor. LKY-047 was found to be a selective CYP2J2 inhibitor with no inhibitory effect on other human P450s, such as CYPs 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A (IC50 > 50 µM). These in vitro data support the use of LKY-047 as a selective CYP2J2 inhibitor with potential application in the identification of P450 isoforms responsible for drug metabolism in reaction phenotyping assays.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 CYP2J2 , Humans , Hydroxylation/drug effects , Inactivation, Metabolic/drug effects , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , NADP/metabolism , Protein Isoforms/metabolism
4.
Bioorg Med Chem Lett ; 26(15): 3529-32, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27329797

ABSTRACT

We synthesized (+)-decursin derivatives substituted with cinnamoyl- and phenyl propionyl groups originating from (+)-CGK062 and screened them using a cell-based assay to detect relative luciferase reporter activity. Of this series, compound 8b, in which a 3-acetoxy cinnamoyl group was introduced, most potently inhibited (97.0%) the Wnt/ß-catenin pathway. Specifically, compound 8b dose-dependently inhibited Wnt3a-induced expression of the ß-catenin response transcription (CRT) and increased ß-catenin degradation in HEK293 reporter cells. Furthermore, compound 8b suppressed expression of the downstream ß-catenin target genes cyclin D1 and c-myc and suppressed PC3 cell growth in a concentration-dependent manner.


Subject(s)
Benzopyrans/pharmacology , Butyrates/pharmacology , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Benzopyrans/chemical synthesis , Benzopyrans/chemistry , Butyrates/chemical synthesis , Butyrates/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Stereoisomerism , Structure-Activity Relationship , Wnt Proteins/metabolism , beta Catenin/metabolism
5.
Bioorg Med Chem Lett ; 25(19): 4304-7, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26271589

ABSTRACT

In this Letter, we investigated the barrier protective effects of 3-N-(MeO)n-cinnamoyl carbazoles (BS 1; n=1, BS 2; n=2, BS 3; n=3) and 3-O-(MeO)3-cinnamoyl carbazole (BS 4) against high-mobility group box 1 (HMGB1)-mediated vascular disruptive responses in human umbilical vein endothelial cells (HUVECs) and in mice for the first time. Data showed that BS 2, BS 3, and BS 4, but not BS 1, inhibited HMGB1-mediated vascular disruptive responses and transendothelial migration of human neutrophils to HUVECs. BS 2, BS3, and BS 4 also suppressed HMGB1-induced hyperpermeability and leukocyte migration in mice. Interestingly, the barrier protective effects of BS 3 and BS 4 were better than those of BS 2. These results suggest that the number of methoxy groups substituted on the cinnamamide or cinnamate moiety of the 9H-3-carbazole derivative is an important pharmacophore for the barrier protective effects of these compounds.


Subject(s)
Carbazoles/chemistry , Carbazoles/pharmacology , HMGB1 Protein/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Leukocytes/drug effects , Animals , Carbazoles/chemical synthesis , Cell Movement/drug effects , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukocytes/metabolism , Mice , Molecular Structure , Permeability/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...