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.
Bioorg Med Chem ; 25(21): 5995-6006, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28988629

ABSTRACT

The discovery of a novel series of ß-methyltryptophan (ß MeTrp) derivatives as selective and orally active non-peptide somatostatin receptor 2 (SSTR2) agonists for the treatment of Type 2 diabetes is described. In our previous research, Compound A, ß-MeTrp derivative with highly potent and selective SSTR2 agonistic activity IC50 (SSTR2/SSTR5)=0.3/>100 (nM), was identified asa drug candidate for treatment of Type 2 diabetes which lowers significantly plasma glucose level in Wistar fatty rats in its oral administrations. However, as serious increase in AUC and phospholipidosis (PLsis) were observed in its toxicological studies in rats, follow-up compounds were searched to avoid risk of PLsis with reference to their in vitro PLsis potentials evaluated on the basis of accumulation of phospholipids in HepG2 cells exposed to the compounds. It has been found that introduction of a carbonyl group onto the piperidine and piperazine or aniline moiety of compounds A and B reduced markedly the in vitro PLsis potentials. And further modification of the compounds and their evaluation led to a discovery of compounds 3k with lower in vitro PLsis potentials exhibiting lowering effect of hypoglycemia-induced glucagon secretion in SD rats (ED50=1.1mg/kg) and glucose excursion in meal tolerance test in Wistar fatty diabetic rats (MED=3.0mg/kg) in oral administrations. Compound 3k was selected asa new drug candidate of selective and orally active non-peptide SSTR2 agonists for treatment of Type 2 diabetes with low in vivo PLsis potential.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Design , Receptors, Somatostatin/agonists , Tryptophan/analogs & derivatives , Administration, Oral , Animals , Dose-Response Relationship, Drug , Humans , Male , Molecular Structure , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tryptophan/administration & dosage , Tryptophan/chemistry , Tryptophan/pharmacology
2.
Toxicol Mech Methods ; 19(8): 477-85, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19793005

ABSTRACT

Drug candidates under development by industry frequently show phospholipidosis as a side-effect in pre-clinical toxicity studies. This study sets up a cell-based assay for drug-induced phospholipidosis (PLD) and its performance was evaluated based on the in vivo PLD potential of compounds in 2-week toxicity studies in rats. When HepG2 cells were exposed simultaneously to PLD-inducing chemicals and a phospholipid having a fluorophore, an accumulation of phospholipids was detected as an increasing fluorescent intensity. Amiodarone, amitriptyline, fluoxetine, AY-9944, and perhexiline, which are common PLD-inducing chemicals, increased the fluorescent intensity, but acetaminophen, ampicillin, cimetidine, famotidine, or valproic acid, which are non-PLD-inducing chemicals, did not. The fluorescent intensity showed concordance with the pathological observations of phospholipid lamellar bodies in the cells. Then to confirm the predictive performance of the in vitro PLD assay, the 32 proprietary compounds characterized in 2-week toxicity studies in rats were evaluated with this in vitro assay. Because this in vitro assay was vulnerable to cytotoxicity, the innate PLD potential was calculated for each compound. A statistically significant increase in the in vitro PLD potential was seen for the compounds having in vivo PLD-inducing potential in the rat toxicity studies. The results suggest that the in vitro PLD potential could be appropriate to detect the appearance of PLD as a side effect in pre-clinical toxicity studies in rats.


Subject(s)
Fluorescent Dyes/metabolism , Lipidoses/chemically induced , Phosphatidylethanolamines/metabolism , Phospholipids/metabolism , Toxicity Tests/methods , Amiodarone/toxicity , Animal Testing Alternatives/methods , Animals , Cytoplasmic Vesicles/drug effects , Enzyme Inhibitors/toxicity , Female , Hep G2 Cells , Humans , Lipidoses/pathology , Male , Microscopy, Electron, Transmission , Organ Specificity/drug effects , Rats , Rats, Sprague-Dawley , Spectrometry, Fluorescence , Time Factors , Toxicity Tests/economics
3.
Neuroreport ; 20(8): 745-9, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19352207

ABSTRACT

Although moderate hypothermia is one of the most robust and effective techniques available for reducing ischemic injury, its key mechanism still remains unclear. Our proteomic analysis of the brains of rats treated with a 2-h middle cerebral artery occlusion showed that postischemic hypothermia markedly potentiated a sustained increase in heat-shock protein 70 (Hsp70). The elevated Hsp70 level was confirmed by enzyme-linked immunosorbent assay, western blot analysis, and immunohistochemical staining. Expression of other Hsp proteins was unaffected by hypothermia. Interestingly, hypothermia did not increased, even decreased, the upregulation of hsp70 mRNA expression by ischemia, suggesting that Hsp70 abundance is controlled by an unknown posttranscriptional regulation. As Hsp70 exerts a protective role against ischemic damage, the specific increase in Hsp70 production may contribute to the neuroprotective effect of hypothermia.


Subject(s)
Brain Infarction/therapy , Brain Ischemia/metabolism , Brain Ischemia/therapy , Cytoprotection/physiology , HSP70 Heat-Shock Proteins/genetics , Hypothermia, Induced/methods , Animals , Biomarkers/analysis , Biomarkers/metabolism , Body Temperature/physiology , Brain/blood supply , Brain/metabolism , Brain/physiopathology , Brain Infarction/metabolism , Brain Infarction/physiopathology , Brain Ischemia/physiopathology , Cell Survival/physiology , Male , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Nerve Degeneration/therapy , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/physiology
4.
Mol Cell Biol ; 28(3): 1171-81, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18039846

ABSTRACT

Regulation of chromatin in eukaryotic transcription requires histone-modifying enzymes, nucleosome remodeling complexes, and histone chaperones. Specific regulation of histone incorporation/eviction by histone chaperones on the promoter (e.g., region specific) is still poorly understood. In the present study, we show that direct and functional interaction of histone chaperone and DNA-binding transcription factor leads to promoter region-specific histone incorporation and inhibition of histone acetylation. We report here that the DNA-binding transcription factor Krüppel-like factor 5 (KLF5) interacts with the novel histone chaperone acidic nuclear phosphoprotein 32B (ANP32B), leading to transcriptional repression of a KLF5-downstream gene. We further show that recruitment of ANP32B onto the promoter region requires KLF5 and results in promoter region-specific histone incorporation and inhibition of histone acetylation by ANP32B. Extracellular stimulus (e.g., phorbol ester) regulates this mechanism in the cell. Collectively, we have identified a novel histone chaperone, ANP32B, and through analysis of the actions of this factor show a new mechanism of promoter region-specific transcriptional regulation at the chromatin level as mediated by the functional interaction between histone chaperone and DNA-binding transcription factor.


Subject(s)
Histones/metabolism , Kruppel-Like Transcription Factors/physiology , Nuclear Proteins/physiology , Promoter Regions, Genetic , Transcription, Genetic , Acetylation , DNA-Binding Proteins/physiology , Gene Expression Regulation , HeLa Cells , Humans , Molecular Chaperones/physiology
5.
Mol Cell Biol ; 23(23): 8528-41, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14612398

ABSTRACT

Here we show a novel pathway of transcriptional regulation of a DNA-binding transcription factor by coupled interaction and modification (e.g., acetylation) through the DNA-binding domain (DBD). The oncogenic regulator SET was isolated by affinity purification of factors interacting with the DBD of the cardiovascular transcription factor KLF5. SET negatively regulated KLF5 DNA binding, transactivation, and cell-proliferative activities. Down-regulation of the negative regulator SET was seen in response to KLF5-mediated gene activation. The coactivator/acetylase p300, on the other hand, interacted with and acetylated KLF5 DBD, and activated its transcription. Interestingly, SET inhibited KLF5 acetylation, and a nonacetylated mutant of KLF5 showed reduced transcriptional activation and cell growth complementary to the actions of SET. These findings suggest a new pathway for regulation of a DNA-binding transcription factor on the DBD through interaction and coupled acetylation by two opposing regulatory factors of a coactivator/acetylase and a negative cofactor harboring activity to inhibit acetylation.


Subject(s)
Nuclear Proteins/metabolism , Proteins/metabolism , Trans-Activators/metabolism , Acetylation , Animals , Binding Sites , Cell Line , Chromosomal Proteins, Non-Histone , DNA, Complementary/genetics , DNA-Binding Proteins , Down-Regulation , E1A-Associated p300 Protein , HeLa Cells , Histone Chaperones , Humans , In Vitro Techniques , Kruppel-Like Transcription Factors , Mice , Models, Biological , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Rabbits , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors , Up-Regulation
6.
J Biol Chem ; 278(31): 28758-64, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12759364

ABSTRACT

Transcription involves molecular interactions between general and regulatory transcription factors with further regulation by protein-protein interactions (e.g. transcriptional cofactors). Here we describe functional interaction between DNA-binding transcription factor and histone chaperone. Affinity purification of factors interacting with the DNA-binding domain of the transcription factor Sp1 showed Sp1 to interact with the histone chaperone TAF-I, both alpha and beta isoforms. This interaction was specific as Sp1 did not interact with another histone chaperone CIA nor did other tested DNA-binding regulatory factors (MyoD, NFkappaB, p53) interact with TAF-I. Interaction of Sp1 and TAF-I occurs both in vitro and in vivo. Interaction with TAF-I results in inhibition of DNA-binding, and also likely as a result of such, inhibition of promoter activation by Sp1. Collectively, we describe interaction between DNA-binding transcription factor and histone chaperone which results in negative regulation of the former. This novel regulatory interaction advances our understanding of the mechanisms of eukaryotic transcription through DNA-binding regulatory transcription factors by protein-protein interactions, and also shows the DNA-binding domain to mediate important regulatory interactions.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Binding Sites , Blotting, Western , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Glutathione Transferase/genetics , HeLa Cells , Histone Chaperones , Humans , Immunosorbent Techniques , Molecular Sequence Data , Promoter Regions, Genetic , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Sp1 Transcription Factor/chemistry , Sp1 Transcription Factor/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription Factors/chemistry , Transcription Factors/genetics
7.
Nat Med ; 8(8): 856-63, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12101409

ABSTRACT

We recently isolated a Krüppel-like zinc-finger transcription factor 5 (KLF5; also known as BTEB2 and IKLF), which is markedly induced in activated vascular smooth-muscle cells and fibroblasts. Here we describe our analysis of the in vivo function of KLF5 using heterozygous KLF5-knockout mice (Klf5(+/-)). In response to external stress, Klf5(+/-) mice showed diminished levels of arterial-wall thickening, angiogenesis, cardiac hypertrophy and interstitial fibrosis. Also, angiotensin II induced expression of KLF5, which in turn activated platelet-derived growth factor-A (PDGF-A) and transforming growth factor-beta (TGF-beta) expression. In addition, we determined that KLF5 interacted with the retinoic-acid receptor (RAR), that synthetic RAR ligands modulated KLF5 transcriptional activity, and that in vivo administration of RAR ligands affected stress responses in the cardiovascular system in a KLF5-dependent manner. KLF5 thus seems to be a key element linking external stress and cardiovascular remodeling.


Subject(s)
Angiotensin II/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Ventricular Remodeling/physiology , Angiotensin II/pharmacology , Animals , Blood Vessels/cytology , Blood Vessels/drug effects , Blood Vessels/metabolism , Cells, Cultured , Collagen Type IV/metabolism , Dibenzazepines/pharmacology , Digestive System/anatomy & histology , Digestive System Physiological Phenomena , Female , Genes, Reporter , Humans , Kruppel-Like Transcription Factors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/cytology , Myocardium/metabolism , Neoplasm Transplantation , Neovascularization, Physiologic , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Retinoids/antagonists & inhibitors , Trans-Activators/genetics , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...