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 ; 7(1): 8455, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814721

ABSTRACT

Osteoblast differentiation is regulated through the successive activation of signaling molecules by a complex interplay of extracellular signals such as bone morphogenetic protein (BMP) and Wnt ligands. Numerous studies have identified natural as well as synthetic compounds with osteogenic activity through the regulation of either BMP/SMADs or the Wnt/ß-catenin pathway. Here, we attempted to isolate small molecules that concurrently activated both SMADs and ß-catenin, which led to the discovery of a novel potent osteogenic compound, DMP-PYT. Upon BMP2 stimulation, DMP-PYT substantially increased osteoblast differentiation featured by enhanced expression of osteoblast-specific genes and accelerated calcification through activation of BMPs expression. DMP-PYT promoted BMP2-induced SMAD1/5/8 phosphorylation and ß-catenin expression, the latter in a BMP2-independent manner. DMP-PYT alone enhanced nuclear localization of ß-catenin to promote the DNA-binding and transcriptional activity of T-cell factor, thereby resulting in increased osteoblast differentiation in the absence of BMP2. Most importantly, DMP-PYT advanced skeletal development and bone calcification in zebrafish larvae. Conclusively, DMP-PYT strongly stimulated osteoblast differentiation and bone formation in vitro and in vivo by potentiating BMP2-induced activation of SMADs and ß-catenin. These results suggest that DMP-PYT may have beneficial effects for preventing and for treating osteoporosis.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , Wnt Proteins/metabolism , Animals , Animals, Genetically Modified , Bone Morphogenetic Proteins/genetics , Cell Differentiation/genetics , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression/drug effects , Larva/genetics , Larva/metabolism , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Molecular Structure , Osteoblasts/cytology , Osteogenesis/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Small Molecule Libraries/chemistry , Wnt Proteins/genetics , Zebrafish/genetics , Zebrafish/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Red Fluorescent Protein
2.
Biochem Biophys Res Commun ; 487(2): 409-414, 2017 05 27.
Article in English | MEDLINE | ID: mdl-28419840

ABSTRACT

Bone mass is controlled by a balance between bone resorption and formation by osteoclasts and osteoblasts, respectively. An imbalance between osteoblasts and osteoclasts increases the risk of osteoporosis and fractures. Although inhibition of osteoclasts is beneficial for preventing and treating osteoporosis, enhanced bone formation through activation of osteoblast differentiation can be a more promising therapeutic approach. In this study, we attempted to isolate small molecules that promote osteoblast differentiation and found that IBIP (3-(2,3-dimethoxyphenyl)-1-[9-methyl-2-phenyl-9H-imidazo[1,2-a]benzimidazol-3-yl]-2-propen-1-one) was a potent activator of osteoblast differentiation. Upon bone morphogenetic protein-2 (BMP2) stimulation, IBIP promoted osteoblast differentiation and increased the expression of osteoblast-specific gene markers, such as osterix and alkaline phosphatase, in a dose-dependent manner. The phosphorylation of SMADs and extracellular signal-regulated kinase (ERK) increased after IBIP treatment. While enhanced SMAD phosphorylation by IBIP was abolished by a BMP inhibitor, IBIP-induced ERK phosphorylation was sustained in the presence of this inhibitor, but was decreased by an ERK kinase inhibitor. Suppression of IBIP-induced SMAD and ERK phosphorylation diminished osteoblast differentiation. Most importantly, IBIP enhanced bone formation and calcification in a BMP2-independent manner in vitro and advanced the skeletal development of zebrafish larvae in vivo. Collectively, IBIP may have beneficial effects on bone loss through potentiation of bone formation.


Subject(s)
Benzimidazoles/administration & dosage , Bone Development/physiology , Osteoblasts/drug effects , Osteoblasts/physiology , Osteogenesis/physiology , 3T3 Cells , Animals , Bone Development/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Imidazoles/administration & dosage , Mice , Osteoblasts/cytology , Osteogenesis/drug effects , Zebrafish
3.
Chem Biodivers ; 11(6): 962-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24934681

ABSTRACT

Antofine (ANTF) is a phenanthroindolizidine alkaloid isolated from the root of Cynanchum paniculatum Kitagawa (Asclepiadaceae), which is used as an herbal remedy for pain and inflammation. ANTF also possesses antiviral and antitumorigenic activities. In this study, we investigated the role of ANTF in adipogenesis. Chronic ABTF administration suppressed adipocyte differentiation and marker expression in a dose-dependent manner. Furthermore, acute administration of ANTF at early stages of differentiation process inhibited lipid droplet formation and adipogenic gene expression. ANTF Treatment decreased expression of PPARγ protein, a master transcription factor in the regulation of adipocyte differentiation, leading to a suppression of aP2 promoter activity. These results suggest that ANTF exerts potent anti-adipogenic effects via direct suppression of PPARγ protein expression, with consequent downregulation of adipogenic gene expression.


Subject(s)
Adipogenesis/drug effects , Biological Products/pharmacology , Indoles/pharmacology , PPAR gamma/biosynthesis , Phenanthrolines/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Adipogenesis/genetics , Animals , Apocynaceae/chemistry , Biological Products/chemistry , Biological Products/isolation & purification , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Indoles/chemistry , Indoles/isolation & purification , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Phenanthrolines/chemistry , Phenanthrolines/isolation & purification , Plant Roots/chemistry , Structure-Activity Relationship
4.
J Mol Endocrinol ; 52(2): 191-202, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24444497

ABSTRACT

Selective inhibitors of 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) have considerable potential as treatment for osteoporosis as well as metabolic syndrome including type 2 diabetes mellitus. Here, we investigated the anti-diabetic, anti-adipogenic, and anti-osteoporotic activity of KR-67500, as a novel selective 11ß-HSD1 inhibitor. Cellular 11ß-HSD1 activity was tested based on a homogeneous time-resolved fluorescence method. Oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) levels were measured in diet-induced obese (DIO)-C57BL/6 mice administered KR-67500 (50  mg/kg per day, p.o.) for 28 days and, additionally, its anti-diabetic effect was evaluated by OGTT and ITT. The in vitro anti-adipogenic effect of KR-67500 was determined by Oil Red O Staining. The in vitro anti-osteoporotic activity of KR-67500 was evaluated using bone morphogenetic protein 2 (BMP2)-induced osteoblast differentiation and receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation model systems. KR-67500 improved the in vivo glucose tolerance and insulin sensitivity in DIO-C57BL/6 mice. KR-67500 suppressed cortisone-induced differentiation of 3T3-L1 cells into adipocytes. KR-67500 enhanced BMP2-induced osteoblastogenesis in C2C12 cells and inhibited RANKL-induced osteoclastogenesis in mouse bone marrow-derived macrophages. KR-67500, a new selective 11ß-HSD1 inhibitor, may provide a new therapeutic window in the prevention and/or treatment of type 2 diabetes, obesity, and/or osteoporosis.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/pathology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Obesity/drug therapy , Osteoblasts/pathology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 3T3-L1 Cells , Adipogenesis/drug effects , Animals , Bone Marrow Cells/pathology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diet , Glucose Tolerance Test , Insulin Resistance , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/pathology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Real-Time Polymerase Chain Reaction
5.
Bioorg Med Chem Lett ; 22(17): 5470-4, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22832316

ABSTRACT

Androgen receptor (AR) is crucial for transcriptional signaling in prostate cancers. The anti-cancer activity of protein kinase CK2 (formerly called casein kinase 2)-specific small molecule inhibitors have been reported in several cancers including prostate cancers. The orally available CX4945, a potent and selective small molecule inhibitor of CK2, has advanced into human clinical trials and has exhibited strong anti-tumor activity. The inhibition of CK2 leads to a down-regulation of the AR-dependent transcription, but the functional relevance of CX4945 to AR-dependent transcription in AR-positive LNCap cells has not been studied yet. Our observation of inhibitory effects of CX4945 on the expression or phosphorylation levels of CK2α, Akt and anti-apoptotic molecules including IAP family members agreed with a previous study showing the effect of CK2 inhibition in cancer cells. This study also provides novel information on the impact of CX4945 in the inhibition of AR-dependent transcriptional activation in LNCap cells via its down-regulation. Pharmacologic inhibition experiment revealed that CX4945 could exhibit its anti-cancer activity in LNCap cells via the independent inhibitions of AR and Akt-survivin signalings.


Subject(s)
Androgen Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Casein Kinase II/antagonists & inhibitors , Naphthyridines/pharmacology , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Humans , Male , Phenazines , Prostatic Neoplasms/metabolism
6.
Amino Acids ; 42(4): 1455-65, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21468757

ABSTRACT

We investigated the in vitro and in vivo osteogenic activity of licochalcone A. At low concentrations, licochalcone A stimulated the differentiation of mouse pre-osteoblastic MC3T3-E1 subclone 4 (MC4) cells and enhanced the bone morphogenetic protein (BMP)-2-induced stimulation of mouse bi-potential mesenchymal precursor C2C12 cells to commit to the osteoblast differentiation pathway. This osteogenic activity of licochalcone A was accompanied by the activation of extracellular-signal regulated kinase (ERK). The involvement of ERK was confirmed in a pharmacologic inhibition study. Additionally, noggin (a BMP antagonist) inhibited the osteogenic activity of licochalcone A in C2C12 cells. Licochalcone A also enhanced the BMP-2-stimulated expression of various BMP mRNAs. This suggested that the osteogenic action of licochalcone A in C2C12 cells could be dependent on BMP signaling and/or expression. We then tested the in vivo osteogenic activity of licochalcone A in two independent animal models. Licochalcone A accelerated the rate of skeletal development in zebrafish and enhanced woven bone formation over the periosteum of mouse calvarial bones. In summary, licochalcone A induced osteoblast differentiation with ERK activation in both MC4 and C2C12 cells and it exhibited in vivo osteogenic activity in zebrafish skeletal development and mouse calvarial bone formation. The dual action of licochalcone A in stimulating bone formation and inhibiting bone resorption, as described in a previous study, might be beneficial in treating bone-related disorders.


Subject(s)
Bone Development/drug effects , Cell Differentiation/drug effects , Chalcones/pharmacology , Osteoblasts/cytology , Animals , Bone Morphogenetic Protein 2 , Cell Line , Mice , Mice, Inbred ICR , Osteoblasts/drug effects , Osteoblasts/metabolism , Signal Transduction/drug effects , Zebrafish
7.
Bone ; 50(1): 364-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22108137

ABSTRACT

Kaempferol (KMP) exerts protective effects against both osteoporosis and obesity by regulating cellular activities, but the underlying molecular mechanisms have not been fully elucidated. TAZ (transcriptional coactivator with PDZ-binding motif) modulates both osteoblast and adipocyte differentiation from mesenchymal stem cells by stimulating the activities of RUNX2 (runt-related transcription factor 2) and suppressing the activities of PPARγ (peroxisome proliferator-activated receptor γ). In this study, we investigated the effects of KMP on TAZ regulated osteoblast and adipocyte differentiation. KMP increased the osteoblast differentiation of mesenchymal cells by facilitating the physical interaction between TAZ and RUNX2, thus the increasing transcriptional activities of RUNX2. KMP also enhanced the association of TAZ with PPARγ, thereby suppressing the gene transcription of PPARγ targets and resulting in diminished adipocyte differentiation. Interestingly, the regulatory effects of kaempferol on RUNX2 and PPARγ-mediated transcriptional activity were impaired in TAZ-null mouse embryonic fibroblasts but recovered by restoration of TAZ expression. Our results demonstrate that KMP fortifies TAZ activity, which enhances RUNX2-mediated osteoblast differentiation and suppresses PPARγ-stimulated adipocyte differentiation, indicating the potential of KMP as an effective therapeutic reagent for controlling bone loss and adiposity through TAZ activation.


Subject(s)
Adipogenesis/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Kaempferols/pharmacology , Osteogenesis/drug effects , Animals , Cell Differentiation/drug effects , Cell Line , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/physiology , PPAR gamma/genetics , PPAR gamma/metabolism , Trans-Activators , Transcription Factors , Transcriptional Activation , Transcriptional Coactivator with PDZ-Binding Motif Proteins
8.
Bioorg Med Chem Lett ; 21(2): 727-9, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21195613

ABSTRACT

In order to identify the binding proteins to anti-resorptive 5-chloro-1-(2,6-dimethylpiperidin-1-yl)-N-tosylpentan-1-imine (1), the chemical affinity matrix for the compound 1 (2b) was designed and synthesized. Using 2b-based chemical proteomics, prohibitin was identified as one of strong binding proteins for 2b.


Subject(s)
Amidines/chemistry , Amidines/pharmacology , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacology , Chromatography, Affinity/methods , Repressor Proteins/metabolism , Animals , Cell Line , Mice , Prohibitins , Protein Binding , Proteomics/methods , Repressor Proteins/isolation & purification , Sulfinic Acids/chemistry , Sulfinic Acids/pharmacology
9.
J Med Chem ; 53(24): 8760-4, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21114291

ABSTRACT

A novel benzopyran-fused molecular framework 7ai was discovered as a specific inhibitor of RANKL-induced osteoclastogenesis using a cell-based TRAP activity assay from drug-like small-molecule libraries constructed by diversity-oriented synthesis. Its inhibitory activity was confirmed by in vitro evaluations including specific inhibition of RANKL-induced ERK phosphorylation and NF-κB transcriptional activation. 7ai can serve as a specific small-molecule modulator for mechanistic studies of RANKL-induced osteoclast differentiation as well as a potential lead for the development of antiresorptive drugs.


Subject(s)
Benzopyrans/chemical synthesis , Bone Density Conservation Agents/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Osteoclasts/drug effects , RANK Ligand/physiology , Animals , Benzopyrans/chemistry , Benzopyrans/pharmacology , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Crystallography, X-Ray , Extracellular Signal-Regulated MAP Kinases/metabolism , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/pharmacology , High-Throughput Screening Assays , Magnetic Resonance Spectroscopy , Mice , NF-kappa B/metabolism , Osteoclasts/cytology , Phosphorylation , RANK Ligand/pharmacology , Small Molecule Libraries , Stem Cells/cytology , Stem Cells/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL