Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Adv ; 7(21)2021 05.
Article in English | MEDLINE | ID: mdl-34020950

ABSTRACT

Cell division cycle 7 (CDC7), a serine/threonine kinase, plays important roles in DNA replication. We developed a highly specific CDC7 inhibitor, TAK-931, as a clinical cancer therapeutic agent. This study aimed to identify the potential combination partners of TAK-931 for guiding its clinical development strategies. Unbiased high-throughput chemical screening revealed that the highest synergistic antiproliferative effects observed were the combinations of DNA-damaging agents with TAK-931. Functional phosphoproteomic analysis demonstrated that TAK-931 suppressed homologous recombination repair activity, delayed recovery from double-strand breaks, and led to accumulation of DNA damages in the combination. Whole-genome small interfering RNA library screening identified sensitivity-modulating molecules, which propose the experimentally predicted target cancer types for the combination, including pancreatic, esophageal, ovarian, and breast cancers. The efficacy of combination therapy in these cancer types was preclinically confirmed in the corresponding primary-derived xenograft models. Thus, our findings would be helpful to guide the future clinical strategies for TAK-931.


Subject(s)
Neoplasms , Recombinational DNA Repair , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA , DNA Damage , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Protein Serine-Threonine Kinases
2.
PLoS One ; 15(1): e0228212, 2020.
Article in English | MEDLINE | ID: mdl-31990961

ABSTRACT

Acetyl-CoA carboxylase (ACC) catalyzes the rate-limiting step in de novo lipogenesis, which is increased in the livers of patients with nonalcoholic steatohepatitis. GS-0976 (firsocostat), an inhibitor of isoforms ACC1 and ACC2, reduced hepatic steatosis and serum fibrosis biomarkers such as tissue inhibitor of metalloproteinase 1 in patients with nonalcoholic steatohepatitis in a randomized controlled trial, although the impact of this improvement on fibrosis has not fully been evaluated in preclinical models. Here, we used Western diet-fed melanocortin 4 receptor-deficient mice that have similar phenotypes to nonalcoholic steatohepatitis patients including progressively developed hepatic steatosis as well as fibrosis. We evaluated the effects of ACC1/2 inhibition on hepatic fibrosis. After the confirmation of significant hepatic fibrosis with a 13-week pre-feeding, GS-0976 (4 and 16 mg/kg/day) treatment for 9 weeks lowered malonyl-CoA and triglyceride content in the liver and improved steatosis, histologically. Furthermore, GS-0976 reduced the histological area of hepatic fibrosis, hydroxyproline content, mRNA expression level of type I collagen in the liver, and plasma tissue metalloproteinase inhibitor 1, suggesting an improvement of hepatic fibrosis. The treatment with GS-0976 was also accompanied by reductions of plasma ALT and AST levels. These data demonstrate that improvement of hepatic lipid metabolism by ACC1/2 inhibition could be a new option to suppress fibrosis progression as well as to improve hepatic steatosis in nonalcoholic steatohepatitis.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gene Knockout Techniques , Liver Cirrhosis/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Receptor, Melanocortin, Type 4/deficiency , Receptor, Melanocortin, Type 4/genetics , Animals , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Isobutyrates/pharmacology , Isobutyrates/therapeutic use , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Organ Size/drug effects , Oxazoles/pharmacology , Oxazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Triglycerides/metabolism
3.
ACS Med Chem Lett ; 10(10): 1498-1503, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31620240

ABSTRACT

General control nonderepressible 2 (GCN2) is a master regulator kinase of amino acid homeostasis and important for cancer survival in the tumor microenvironment under amino acid depletion. We initiated studies aiming at the discovery of novel GCN2 inhibitors as first-in-class antitumor agents and conducted modification of the substructure of sulfonamide derivatives with expected type I half binding on GCN2. Our synthetic strategy mainly corresponding to the αC-helix allosteric pocket of GCN2 led to significant enhancement in potency and a good pharmacokinetic profile in mice. In addition, compound 6d, which showed slow dissociation in binding on GCN2, demonstrated antiproliferative activity in combination with the asparagine-depleting agent asparaginase in an acute lymphoblastic leukemia (ALL) cell line, and it also displayed suppression of GCN2 pathway activation with asparaginase treatment in the ALL cell line and mouse xenograft model.

4.
Sci Adv ; 5(5): eaav3660, 2019 05.
Article in English | MEDLINE | ID: mdl-31131319

ABSTRACT

Replication stress (RS) is a cancer hallmark; chemotherapeutic drugs targeting RS are widely used as treatments for various cancers. To develop next-generation RS-inducing anticancer drugs, cell division cycle 7 (CDC7) has recently attracted attention as a target. We have developed an oral CDC7-selective inhibitor, TAK-931, as a candidate clinical anticancer drug. TAK-931 induced S phase delay and RS. TAK-931-induced RS caused mitotic aberrations through centrosome dysregulation and chromosome missegregation, resulting in irreversible antiproliferative effects in cancer cells. TAK-931 exhibited significant antiproliferative activity in preclinical animal models. Furthermore, in indication-seeking studies using large-scale cell panel data, TAK-931 exhibited higher antiproliferative activities in RAS-mutant versus RAS-wild-type cells; this finding was confirmed in pancreatic patient-derived xenografts. Comparison analysis of cell panel data also demonstrated a unique efficacy spectrum for TAK-931 compared with currently used chemotherapeutic drugs. Our findings help to elucidate the molecular mechanisms for TAK-931 and identify potential target indications.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazolones/pharmacology , Pyrimidines/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation , Cell Separation , Cell Survival , Centrosome/drug effects , Chromosome Aberrations/drug effects , Computational Biology , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Female , HeLa Cells , Humans , Inhibitory Concentration 50 , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mitosis/drug effects , Models, Animal , Mutation , Neoplasm Transplantation , Pancreatic Neoplasms/drug therapy , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proteomics , Treatment Outcome , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 115(33): E7776-E7785, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061420

ABSTRACT

General control nonderepressible 2 (GCN2) plays a major role in the cellular response to amino acid limitation. Although maintenance of amino acid homeostasis is critical for tumor growth, the contribution of GCN2 to cancer cell survival and proliferation is poorly understood. In this study, we generated GCN2 inhibitors and demonstrated that inhibition of GCN2 sensitizes cancer cells with low basal-level expression of asparagine synthetase (ASNS) to the antileukemic agent l-asparaginase (ASNase) in vitro and in vivo. We first tested acute lymphoblastic leukemia (ALL) cells and showed that treatment with GCN2 inhibitors rendered ALL cells sensitive to ASNase by preventing the induction of ASNS, resulting in reduced levels of de novo protein synthesis. Comprehensive gene-expression profiling revealed that combined treatment with ASNase and GCN2 inhibitors induced the stress-activated MAPK pathway, thereby triggering apoptosis. By using cell-panel analyses, we also showed that acute myelogenous leukemia and pancreatic cancer cells were highly sensitive to the combined treatment. Notably, basal ASNS expression at protein levels was significantly correlated with sensitivity to combined treatment. These results provide mechanistic insights into the role of GCN2 in the amino acid response and a rationale for further investigation of GCN2 inhibitors for the treatment of cancer.


Subject(s)
Amino Acids/metabolism , Asparaginase/pharmacology , Aspartate-Ammonia Ligase/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amino Acids/genetics , Aspartate-Ammonia Ligase/genetics , Cell Line, Tumor , Humans , Neoplasm Proteins/genetics , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
6.
Bioorg Med Chem Lett ; 26(17): 4296-300, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27476141

ABSTRACT

Centromere-associated protein-E (CENP-E) is a mitotic kinesin which plays roles in cell division, and is regarded as a promising therapeutic target for the next generation of anti-mitotic agents. We designed novel fused bicyclic CENP-E inhibitors starting from previous reported dihydrobenzofuran derivative (S)-(+)-1. Our design concept was to adjust the electron density distribution on the benzene ring of the dihydrobenzofuran moiety to increase the positive charge for targeting the negatively charged L5 loop of CENP-E, using predictions from electrostatic potential map (EPM) analysis. For the efficient synthesis of our 2,3-dihydro-1-benzothiophene 1,1-dioxide derivatives, a new synthetic method was developed. As a result, we discovered 6-cyano-7-trifluoromethyl-2,3-dihydro-1-benzothiophene 1,1-dioxide derivative (+)-5d (Compound A) as a potent CENP-E inhibitor with promising potential for in vivo activity. In this Letter, we discuss the design and synthetic strategy used in the discovery of (+)-5d and structure-activity relationships for its analogs possessing various fused bicyclic L5 binding moieties.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Cyclic S-Oxides/chemical synthesis , Drug Delivery Systems , Drug Design , Imidazoles/chemical synthesis , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclic S-Oxides/chemistry , Cyclic S-Oxides/pharmacology , HeLa Cells , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Inhibitory Concentration 50 , Molecular Structure , Protein Binding/drug effects , Structure-Activity Relationship
7.
PLoS One ; 10(12): e0144675, 2015.
Article in English | MEDLINE | ID: mdl-26649895

ABSTRACT

Centromere-associated protein E (CENP-E) regulates both chromosome congression and the spindle assembly checkpoint (SAC) during mitosis. The loss of CENP-E function causes chromosome misalignment, leading to SAC activation and apoptosis during prolonged mitotic arrest. Here, we describe the biological and antiproliferative activities of a novel small-molecule inhibitor of CENP-E, Compound-A (Cmpd-A). Cmpd-A inhibits the ATPase activity of the CENP-E motor domain, acting as a time-dependent inhibitor with an ATP-competitive-like behavior. Cmpd-A causes chromosome misalignment on the metaphase plate, leading to prolonged mitotic arrest. Treatment with Cmpd-A induces antiproliferation in multiple cancer cell lines. Furthermore, Cmpd-A exhibits antitumor activity in a nude mouse xenograft model, and this antitumor activity is accompanied by the elevation of phosphohistone H3 levels in tumors. These findings demonstrate the potency of the CENP-E inhibitor Cmpd-A and its potential as an anticancer therapeutic agent.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Spindle Apparatus/drug effects , Animals , Cell Line, Tumor , Chromosome Segregation , Heterografts , Metaphase , Mice , Mice, Nude , Microtubules/metabolism , Mitosis
8.
Nat Commun ; 6: 7668, 2015 Jul 06.
Article in English | MEDLINE | ID: mdl-26144554

ABSTRACT

The molecular mechanism responsible that determines cell fate after mitotic slippage is unclear. Here we investigate the post-mitotic effects of different mitotic aberrations--misaligned chromosomes produced by CENP-E inhibition and monopolar spindles resulting from Eg5 inhibition. Eg5 inhibition in cells with an impaired spindle assembly checkpoint (SAC) induces polyploidy through cytokinesis failure without a strong anti-proliferative effect. In contrast, CENP-E inhibition causes p53-mediated post-mitotic apoptosis triggered by chromosome missegregation. Pharmacological studies reveal that aneuploidy caused by the CENP-E inhibitor, Compound-A, in SAC-attenuated cells causes substantial proteotoxic stress and DNA damage. Polyploidy caused by the Eg5 inhibitor does not produce this effect. Furthermore, p53-mediated post-mitotic apoptosis is accompanied by aneuploidy-associated DNA damage response and unfolded protein response activation. Because Compound-A causes p53 accumulation and antitumour activity in an SAC-impaired xenograft model, CENP-E inhibitors could be potential anticancer drugs effective against SAC-impaired tumours.


Subject(s)
Aneuploidy , Apoptosis , DNA Damage , M Phase Cell Cycle Checkpoints/physiology , Tumor Suppressor Protein p53/physiology , Animals , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , HeLa Cells , Heterografts , Humans , Kinesins/antagonists & inhibitors , Mice , Mice, Nude , Mitosis , Neoplasms, Experimental , Stress, Physiological
9.
Life Sci ; 89(21-22): 765-72, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-21971119

ABSTRACT

AIMS: GPR61 is an orphan G protein-coupled receptor whose function remains unknown. The purpose of the present study is to elucidate the importance of GPR61 in metabolism by characterization of GPR61-deficient mice. MAIN METHODS: Male GPR61-deficient mice were characterized regarding various metabolic parameters, including food intake, body weight, oxygen consumption, body temperature, locomotor activity, and in a pair feeding study. Hypothalamic gene expression was analyzed using real-time quantitative RT-PCR. KEY FINDINGS: GPR61-deficient mice exhibited marked hyperphagia and heavier body weight than wild-type mice. Hyperphagia of GPR61-deficient mice was observed before the differences in body weight became apparent between the genotypes. When body weight difference did become apparent between genotypes, increases in visceral fat pad weight, liver weight, liver triglyceride (TG) content, plasma leptin, and plasma insulin were observed in GPR61-deficient mice, suggesting that GPR61 deficiency caused obesity associated with hyperphagia. Oxygen consumption, body temperature, and locomotor activity were not significantly different between GPR61-deficient and wild-type mice. Pair-fed GPR61-deficient mice had a greater fat mass than wild-type mice despite comparable body weight in both genotypes. The mRNA levels of proopiomelanocortin (POMC) and brain-derived neurotropic factor (BDNF) in the hypothalamus of GPR61-deficient mice were significantly lower than those of wild-type mice. SIGNIFICANCE: GPR61-deficient mice exhibited obesity associated with hyperphagia. These findings suggest that GPR61 is involved in the regulation of food intake and body weight, and may be of importance when considering GPR61 as a therapeutic target for obesity or eating disorders.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Animals , Body Composition/genetics , Body Temperature/genetics , Body Weight/genetics , Calorimetry, Indirect , Diet , Eating/genetics , Galactosides , Immunohistochemistry , Indoles , Lac Operon , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Organ Size/genetics , Oxygen Consumption/drug effects , Phenotype , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Triglycerides/metabolism
10.
Mol Cancer Ther ; 9(1): 157-66, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20053775

ABSTRACT

Aurora-A kinase is a one of the key regulators during mitosis progression. Aurora-A kinase is a potential target for anticancer therapies because overexpression of Aurora-A, which is frequently observed in some human cancers, results in aberrant mitosis leading to chromosomal instability and possibly tumorigenesis. MK-5108 is a novel small molecule with potent inhibitory activity against Aurora-A kinase. Although most of the Aurora-kinase inhibitors target both Aurora-A and Aurora-B, MK-5108 specifically inhibited Aurora-A kinase in a panel of protein kinase assays. Inhibition of Aurora-A by MK-5108 in cultured cells induced cell cycle arrest at the G(2)-M phase in flow cytometry analysis. The effect was confirmed by the accumulation of cells with expression of phosphorylated Histone H3 and inhibition of Aurora-A autophosphorylation by immunostaining assays. MK-5108 also induced phosphorylated Histone H3 in skin and xenograft tumor tissues in a nude rat xenograft model. MK-5108 inhibited growth of human tumor cell lines in culture and in different xenograft models. Furthermore, the combination of MK-5108 and docetaxel showed enhanced antitumor activities compared with control and docetaxel alone-treated animals without exacerbating the adverse effects of docetaxel. MK-5108 is currently tested in clinical trials and offers a new therapeutic approach to combat human cancers as a single agent or in combination with existing taxane therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Taxoids/pharmacology , Thiazoles/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Aurora Kinase A , Aurora Kinase B , Aurora Kinases , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclohexanecarboxylic Acids/administration & dosage , Cyclohexanecarboxylic Acids/chemistry , Docetaxel , Humans , Inhibitory Concentration 50 , Mice , Mitosis/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Rats , Taxoids/toxicity , Thiazoles/administration & dosage , Thiazoles/chemistry , Xenograft Model Antitumor Assays
11.
Mol Cancer Ther ; 8(11): 2992-3000, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19887545

ABSTRACT

Wee1 is a tyrosine kinase that phosphorylates and inactivates CDC2 and is involved in G(2) checkpoint signaling. Because p53 is a key regulator in the G(1) checkpoint, p53-deficient tumors rely only on the G(2) checkpoint after DNA damage. Hence, such tumors are selectively sensitized to DNA-damaging agents by Wee1 inhibition. Here, we report the discovery of a potent and selective small-molecule inhibitor of Wee1 kinase, MK-1775. This compound inhibits phosphorylation of CDC2 at Tyr15 (CDC2Y15), a direct substrate of Wee1 kinase in cells. MK-1775 abrogates G(2) DNA damage checkpoint, leading to apoptosis in combination with DNA-damaging chemotherapeutic agents such as gemcitabine, carboplatin, and cisplatin selectively in p53-deficient cells. In vivo, MK-1775 potentiates tumor growth inhibition by these agents, and cotreatment does not significantly increase toxicity. The enhancement of antitumor effect by MK-1775 was well correlated with inhibition of CDC2Y15 phosphorylation in tumor tissue and skin hair follicles. Our data indicate that Wee1 inhibition provides a new approach for treatment of multiple human malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , DNA Damage , Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Tumor Suppressor Protein p53/deficiency , Animals , Apoptosis/drug effects , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin B/metabolism , Cyclin-Dependent Kinases , Drug Synergism , Flow Cytometry , HeLa Cells , Humans , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Pyrimidinones , Rats , Rats, Inbred F344 , Rats, Nude , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
12.
Biochem Biophys Res Commun ; 384(2): 173-9, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19401193

ABSTRACT

Patients with diabetes are under a hypercoagulable state leading to generation of thrombin. It is not known whether thrombin plays a role in the progression of diabetic nephropathy. We analyzed gene expression of two thrombin receptors, protease-activated receptor-1 (PAR-1) and PAR-4 in the kidney of diabetic db/db mice. Mice developed hyperglycemia from 7 to 10 weeks of age and showed renal abnormalities such as mesangial expansion and urinary albumin excretion at 10 weeks of age. PAR-1 mRNA was up-regulated in isolated glomeruli in db/db mice compared with age-matched db/m littermates, but PAR-4 mRNA was not. In situ hybridization studies showed that PAR-1 mRNA was detected mainly at the glomerulus, and that intensive signals were observed in mesangial cells and podocytes. The up-regulation of PAR-1 in glomeruli in diabetic mice may play a role in the progression of glomerulosclerosis and abnormal urinary albumin excretion in diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney Glomerulus/metabolism , Receptor, PAR-1/metabolism , Animals , Diabetes Mellitus/metabolism , Kidney Glomerulus/abnormalities , Mice , Mice, Inbred Strains , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, PAR-1/genetics , Receptors, Thrombin/biosynthesis , Receptors, Thrombin/genetics , Up-Regulation
13.
Endocrinology ; 150(3): 1147-54, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19008318

ABSTRACT

We investigated the effect of glucokinase activator (GKA) on glucose metabolism and beta-cell mass. We analyzed four mouse groups: wild-type mice and beta-cell-specific haploinsufficiency of glucokinase gene (Gck(+/-)) mice on a high-fat (HF) diet. Each genotype was also treated with GKA mixed in the HF diet. Rodent insulinoma cells and isolated islets were used to evaluate beta-cell proliferation by GKA. After 20 wk on the above diets, there were no differences in body weight, lipid profiles, and liver triglyceride content among the four groups. Glucose tolerance was improved shortly after the GKA treatment in both genotypes of mice. beta-Cell mass increased in wild-type mice compared with Gck(+/-) mice, but a further increase was not observed after the administration of GKA in both genotypes. Interestingly, GKA was able to up-regulate insulin receptor substrate-2 (Irs-2) expression in insulinoma cells and isolated islets. The administration of GKA increased 5-bromo-2-deoxyuridine (BrdU) incorporation in insulinoma cells, and 3 d administration of GKA markedly increased BrdU incorporation in mice treated with GKA in both genotypes, compared with those without GKA. In conclusion, GKA was able to chronically improve glucose metabolism for mice on the HF diet. Although chronic GKA administration failed to cause a further increase in beta-cell mass in vivo, GKA was able to increase beta cell proliferation in vitro and with a 3-d administration in vivo. This apparent discrepancy can be explained by a chronic reduction in ambient blood glucose levels by GKA treatment.


Subject(s)
Glucokinase/metabolism , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/cytology , Animals , Body Weight/drug effects , Bromodeoxyuridine/pharmacokinetics , Cells, Cultured , Diet, Atherogenic , Dietary Fats/pharmacology , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Enzyme Activation/physiology , Glucokinase/genetics , Glucose Intolerance/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Mice, Transgenic , Organ Size/drug effects
14.
J Biol Chem ; 279(41): 42867-74, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15292182

ABSTRACT

Intestinal epithelial cells undergo rapid turnover and exfoliation especially at the villus tips. This process is modulated by various nutrients especially fat. Apoptosis is one of the important regulatory mechanisms of this turnover. Therefore, identification of the factors that control epithelial cell apoptosis should help us understand the mechanism of intestinal mucosal turnover. Here, we report the identification of a novel small intestine-specific member of the Ly-6 family, intectin, by signal sequence trap method. Intectin mRNA expression was exclusively identified in the intestine and localized at the villus tips of intestinal mucosa, which is known to undergo apoptosis. Intectin mRNA expression was modulated by nutrition. Intestinal epithelial cells expressing intectin were more sensitive to palmitate-induced apoptosis, compared with control intestinal epithelial cells, and such effect was accompanied by increased activity of caspase-3. Intectin expression also reduced cell-cell adhesion of intestinal epithelial cells.


Subject(s)
Apoptosis , Epithelial Cells/metabolism , Glycosylphosphatidylinositols/chemistry , Intestinal Mucosa/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/physiology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Membrane/metabolism , Cloning, Molecular , Cytosol/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Epithelium/metabolism , Genetic Vectors , In Situ Hybridization , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Palmitic Acid/chemistry , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Subcellular Fractions , Time Factors , Tissue Distribution
15.
J Invest Dermatol ; 122(3): 565-73, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15086536

ABSTRACT

Retinoic acid-inducible gene-1 was originally identified as an orphan G-protein coupled receptor induced by retinoic acid. Three highly homologous oGPCR (GPRC5B, GPRC5C, and GPRC5D) have since been classified into the RAIG1 family. We describe here, the unique tissue distribution of GPRC5D and its mechanism of expression. Hybridization in situ has shown that GPRC5D is expressed in differentiating cells that produce hard keratin, including cortical cells of the hair shaft, the keratogenous zone of the nail, and in a central region of the filiform papillae of the tongue. The GPRC5D transcript is expressed in hair follicles during mid- and late anagen, and catagen but not at telogen and early anagen phases. The differentiation-inducer, all-trans retinoic acid, induces GPRC5D expression in cultured hair bulb cells. Because the tissue distribution of GPRC5D indicates a relationship with hard keratins that constitute the major structural proteins of hard epithelial tissues, we investigated the effect of GPRC5D on acid hard keratins. Analyses of cultured cells showed that transient overexpression resulted in suppression of Ha3 and stimulation of Ha4 hair keratin gene expression. The expression was maintained in the hair follicles of whn-deficient (nude) mice, suggesting that this gene is regulated by a signal pathway different from that of hair keratin synthesis. Collectively, these data provide a framework for understanding the molecular mechanisms of GPRC5D function in hard keratinization.


Subject(s)
Hair/metabolism , Keratins/biosynthesis , Receptors, G-Protein-Coupled/physiology , Animals , Cell Differentiation/drug effects , Hair/chemistry , Hair/cytology , In Situ Hybridization , Keratins/analysis , Mice , Mice, Inbred Strains , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/pharmacology
16.
Eur J Neurosci ; 19(6): 1524-34, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15066149

ABSTRACT

Orexin-A and -B (hypocretin-1 and -2) have been implicated in the stimulation of feeding. Here we show the effector neurons and signaling mechanisms for the orexigenic action of orexins in rats. Immunohistochemical methods showed that orexin axon terminals contact with neuropeptide Y (NPY)- and proopiomelanocortin (POMC)-positive neurons in the arcuate nucleus (ARC) of the rats. Microinjection of orexins into the ARC markedly increased food intake. Orexins increased cytosolic Ca(2+) concentration ([Ca(2+)](i)) in the isolated neurons from the ARC, which were subsequently shown to be immunoreactive for NPY. The increases in [Ca(2+)](i) were inhibited by blockers of phospholipase C (PLC), protein kinase C (PKC) and Ca(2+) uptake into endoplasmic reticulum. The stimulation of food intake and increases in [Ca(2+)](i) in NPY neurons were greater with orexin-A than with orexin-B, indicative of involvement of the orexin-1 receptor (OX(1)R). In contrast, orexin-A and -B equipotently attenuated [Ca(2+)](i) oscillations and decreased [Ca(2+)](i) levels in POMC-containing neurons. These effects were counteracted by pertussis toxin, suggesting involvement of the orexin-2 receptor and Gi/Go subtypes of GTP-binding proteins. Orexins also decreased [Ca(2+)](i) levels in glucose-responsive neurons in the ventromedial hypothalamus (VMH), a satiety center. Leptin exerted opposite effects on these three classes of neurons. These results demonstrate that orexins directly regulate NPY, POMC and glucose-responsive neurons in the ARC and VMH, in a manner reciprocal to leptin. Orexin-A evokes Ca(2+) signaling in NPY neurons via OX(1)R-PLC-PKC and IP(3) pathways. These neural pathways and intracellular signaling mechanisms may play key roles in the orexigenic action of orexins.


Subject(s)
Calcium Signaling/drug effects , Carrier Proteins/pharmacology , Glucose/metabolism , Hypothalamus/cytology , Intracellular Signaling Peptides and Proteins , Leptin/pharmacology , Neuropeptide Y/metabolism , Neuropeptides/pharmacology , Pro-Opiomelanocortin/metabolism , Animals , Dose-Response Relationship, Drug , Drug Interactions , Eating/drug effects , Enzyme Inhibitors , Fura-2/metabolism , Immunohistochemistry , Models, Neurological , Neurons/classification , Neurons/drug effects , Neurons/metabolism , Orexins , Pertussis Toxin/pharmacology , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...