Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Invest New Drugs ; 33(5): 1048-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26076682

ABSTRACT

PURPOSE: The aim of the present study was to assess the safety, maximum tolerated dose (MTD), pharmacokinetics, pharmacodynamics, and efficacy of single and multiple doses of intravenous CG200745, a novel histone deacetylase (HDAC) inhibitor, in patients with advanced solid malignancies. EXPERIMENTAL DESIGN: Two to six patients received intravenous CG200745 according to the 2 + 4 dose-escalating method. This first-in-human trial was comprised of two parts: Part 1 was a single ascending dose, and Part 2 was multiple ascending doses weekly for 3 weeks, and then 1 week off. For the first cycle, pharmacokinetic sampling for CG200745 and pharmacodynamic sampling for acetylated histone H4 in peripheral blood mononuclear cells (PBMCs) were performed on day 1 for Part 1 and on days 1 and 15 for Part 2. Examination of acetylated histone H4 in pre- and post-biopsy samples was performed in accessible patients. RESULTS: In all, 28 patients were treated at 13 dose levels (1.8-250 mg/m(2)) and received a total of 71 cycles of CG200745. Hematologic toxicities included grade 3/4 neutropenia (22.2 %) that did not last a week and non-hematologic toxicities included fatigue (22.2 %) and anorexia (16.7 %) that did not exceed grade 2. No dose-limiting toxic effects were noted. Dose proportionality was observed for both the maximum concentration and area under the curve. The elimination half-life was 5.67 ± 2.69 h (mean ± standard deviation). An increase in PBMC acetylated histone H4 was observed at dose levels up to 51 mg/m(2), which plateaued at higher dose levels. At 24 h, 75 % of patients (6/8) showed higher relative acetylation in tumor tissue compared to PBMCs. Although there was no partial or complete response, 57.1 % of patients (16/28) had stable disease that lasted at least 6 weeks. CONCLUSIONS: CG200745 can be safely administered at effective dose levels that inhibit HDAC in PBMCs and tumor tissue. Although MTD was not reached, further escalation was not performed because acetylated histone H4 plateaued at dose levels higher than 51 mg/m(2). Additional phase II trials are recommended at 250 mg/m(2).


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Neoplasms/drug therapy , Adolescent , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Child , Child, Preschool , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Half-Life , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histones/metabolism , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/pharmacokinetics , Leukocytes, Mononuclear/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Naphthalenes/adverse effects , Naphthalenes/pharmacokinetics , Young Adult
2.
J Biol Chem ; 286(2): 1157-64, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21081500

ABSTRACT

Glucose-6-phosphatase (G6Pase) is a key enzyme that is responsible for the production of glucose in the liver during fasting or in type 2 diabetes mellitus (T2DM). During fasting or in T2DM, peroxisome proliferator-activated receptor α (PPARα) is activated, which may contribute to increased hepatic glucose output. However, the mechanism by which PPARα up-regulates hepatic G6Pase gene expression in these states is not well understood. We evaluated the mechanism by which PPARα up-regulates hepatic G6Pase gene expression in fasting and T2DM states. In PPARα-null mice, both hepatic G6Pase and phosphoenolpyruvate carboxykinase levels were not increased in the fasting state. Moreover, treatment of primary cultured hepatocytes with Wy14,643 or fenofibrate increased the G6Pase mRNA level. In addition, we have localized and characterized a PPAR-responsive element in the promoter region of the G6Pase gene. Chromatin immunoprecipitation (ChIP) assay revealed that PPARα binding to the putative PPAR-responsive element of the G6Pase promoter was increased in fasted wild-type mice and db/db mice. These results indicate that PPARα is responsible for glucose production through the up-regulation of hepatic G6Pase gene expression during fasting or T2DM animal models.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation, Enzymologic , Glucose-6-Phosphatase/genetics , PPAR alpha/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Eating/physiology , Fasting/physiology , Gluconeogenesis/physiology , Hep G2 Cells , Humans , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , PPAR alpha/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Up-Regulation/physiology
3.
Cell Chem Biol ; 25(4): 426-438.e4, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29429898

ABSTRACT

Kynurenine 3-monooxygenase (KMO) inhibitors have been developed for the treatment of neurodegenerative disorders. The mechanisms of flavin reduction and hydrogen peroxide production by KMO inhibitors are unknown. Herein, we report the structure of human KMO and crystal structures of Saccharomyces cerevisiae (sc) and Pseudomonas fluorescens (pf) KMO with Ro 61-8048. Proton transfer in the hydrogen bond network triggers flavin reduction in p-hydroxybenzoate hydroxylase, but the mechanism triggering flavin reduction in KMO is different. Conformational changes via π-π interactions between the loop above the flavin and substrate or non-substrate effectors lead to disorder of the C-terminal α helix in scKMO and shifts of domain III in pfKMO, stimulating flavin reduction. Interestingly, Ro 61-8048 has two different binding modes. It acts as a competitive inhibitor in scKMO and as a non-substrate effector in pfKMO. These findings provide understanding of the catalytic cycle of KMO and insight for structure-based drug design of KMO inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/metabolism , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Kynurenine 3-Monooxygenase/metabolism , Pseudomonas fluorescens/enzymology , Saccharomyces cerevisiae/enzymology , Sulfonamides/pharmacology , Thiazoles/pharmacology , Amino Acid Sequence , Animals , Flavins/metabolism , Humans , Kynurenine 3-Monooxygenase/chemistry , Molecular Docking Simulation , Oxidation-Reduction/drug effects , Protein Conformation/drug effects , Pseudomonas fluorescens/chemistry , Saccharomyces cerevisiae/chemistry , Sequence Alignment
4.
Biochem J ; 399(1): 131-9, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16787385

ABSTRACT

Expression of the GLUT4 (glucose transporter type 4 isoform) gene in adipocytes is subject to hormonal or metabolic control. In the present study, we have characterized an adipose tissue transcription factor that is influenced by fasting/refeeding regimens and insulin. Northern blotting showed that refeeding increased GLUT4 mRNA levels for 24 h in adipose tissue. Consistent with an increased GLUT4 gene expression, the mRNA levels of SREBP (sterol-regulatory-element-binding protein)-1c in adipose tissue were also increased by refeeding. In streptozotocin-induced diabetic rats, insulin treatment increased the mRNA levels of GLUT4 in adipose tissue. Serial deletion, luciferase reporter assays and electrophoretic mobility-shift assay studies indicated that the putative sterol response element is located in the region between bases -109 and -100 of the human GLUT4 promoter. Transduction of the SREBP-1c dominant negative form to differentiated 3T3-L1 adipocytes caused a reduction in the mRNA levels of GLUT4, suggesting that SREBP-1c mediates the transcription of GLUT4. In vivo chromatin immunoprecipitation revealed that refeeding increased the binding of SREBP-1 to the putative sterol-response element in the GLUT4. Furthermore, treating streptozotocin-induced diabetic rats with insulin restored SREBP-1 binding. In addition, we have identified an Sp1 binding site adjacent to the functional sterol-response element in the GLUT4 promoter. The Sp1 site appears to play an additive role in SREBP-1c mediated GLUT4 gene upregulation. These results suggest that upregulation of GLUT4 gene transcription might be directly mediated by SREBP-1c in adipose tissue.


Subject(s)
Adipocytes/metabolism , Gene Expression Regulation , Glucose Transporter Type 4/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cell Differentiation , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Eating , Fasting , Gene Expression Regulation/drug effects , Humans , Insulin/pharmacology , Male , Mice , Promoter Regions, Genetic , Rats , Response Elements/genetics , Sp1 Transcription Factor/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Up-Regulation/drug effects
5.
Sci Rep ; 7: 41615, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28134290

ABSTRACT

Pancreatic cancer is predominantly lethal, and is primarily treated using gemcitabine, with increasing resistance. Therefore, novel agents that increase tumor sensitivity to gemcitabine are needed. Histone deacetylase (HDAC) inhibitors are emerging therapeutic agents, since HDAC plays an important role in cancer initiation and progression. We evaluated the antitumor effect of a novel HDAC inhibitor, CG200745, combined with gemcitabine/erlotinib on pancreatic cancer cells and gemcitabine-resistant pancreatic cancer cells. Three pancreatic cancer-cell lines were used to evaluate the antitumor effect of CG200745 combined with gemcitabine/erlotinib. CG200745 induced the expression of apoptotic proteins (PARP and caspase-3) and increased the levels of acetylated histone H3. CG200745 with gemcitabine/erlotinib showed significant growth inhibition and synergistic antitumor effects in vitro. In vivo, gemcitabine/erlotinib and CG200745 reduced tumor size up to 50%. CG200745 enhanced the sensitivity of gemcitabine-resistant pancreatic cancer cells to gemcitabine, and decreased the level of ATP-binding cassette-transporter genes, especially multidrug resistance protein 3 (MRP3) and MRP4. The novel HDAC inhibitor, CG200745, with gemcitabine/erlotinib had a synergistic anti-tumor effect on pancreatic cancer cells. CG200745 significantly improved pancreatic cancer sensitivity to gemcitabine, with a prominent antitumor effect on gemcitabine-resistant pancreatic cancer cells. Therefore, improved clinical outcome is expected in the future.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Naphthalenes/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Disease Models, Animal , Humans , Mice , Pancreatic Neoplasms/metabolism , Xenograft Model Antitumor Assays , Gemcitabine
6.
IUBMB Life ; 59(3): 134-45, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17487684

ABSTRACT

The gene expression of glucose transporter type 4 isoform (GLUT4) is known to be controlled by metabolic, nutritional, or hormonal status. Understanding the molecular mechanisms governing GLUT4 gene expression is critical, because glucose disposal in the body depends on the activities of GLUT4 in the muscle and adipocytes. The GLUT4 activities are regulated by a variety of mechanisms. One of them is transcriptional regulation. GLUT4 gene expression is regulated by a variety of transcriptional factors in muscle and adipose tissue. These data are accumulating regarding the transcriptional factors regulating GLUT4 gene expression. These include MyoD, MEF2A, GEF, TNF-alpha, TR-1alpha, KLF15, SREBP-1c, C/EBP-alpha, O/E-1, free fatty acids, PAPRgamma, LXRalpha, NF-1, etc. These factors are involved in the positive or negative regulation of GLUT4 gene expression. In addition, there is a complex interplay between these factors in transactivating GLUT4 promoter activity. Understanding the mechanisms controlling GLUT4 gene transcription in these tissues will greatly promote the potential therapeutic drug development for obesity and T2DM.


Subject(s)
Adipocytes/physiology , Gene Expression Regulation , Glucose Transporter Type 4/metabolism , Muscles/physiology , Protein Isoforms/metabolism , Amino Acid Sequence , Animals , Glucose Transporter Type 4/genetics , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Protein Isoforms/genetics , Sequence Alignment , Sequence Analysis, Protein
7.
Biochem Biophys Res Commun ; 360(2): 301-6, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17601490

ABSTRACT

The mechanism of how PPARgamma decrease gluconeogenic gene expressions in liver is still unclear. Since PPARgamma is a transcriptional activator, it requires a mediator to decrease the transcription of gluconeogenic genes. Recently, SHP has been shown to mediate the bile acid-dependent down regulation of gluconeogenic gene expression in liver. This led us to explore the possibility that SHP may mediate the antigluconeogenic effect of PPARgamma. In the present study, we have identified and characterized the presence of functional PPRE in human SHP promoter. We show the binding of PPARgamma/RXRalpha heterodimer to the PPRE and increased SHP expression by rosiglitazone in primary rat hepatocytes. Taken together with the previous reports about the function of SHP on gluconeogenesis, our results indicate that SHP can mediate the acute antigluconeogenic effect of PPARgamma.


Subject(s)
Hepatocytes/metabolism , Liver/metabolism , PPAR gamma/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Transcriptional Activation/physiology , Animals , Cells, Cultured , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL