Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Biophys Res Commun ; 379(4): 861-5, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19126404

ABSTRACT

It is well known that oxidative stress plays critical roles in the pathogenesis of atherosclerosis. In this study, we enrolled 1746 type 2 diabetic subjects, determined 4 common genetic variants related to oxidative stress (glutamate-cysteine ligase modifier subunit (GCLM) C-588T, myeloperoxidase G-463A, human paraoxonase 1 Gln192Arg and NAD(P)H oxidase p22phox C242T polymorphisms), and measured carotid intima-media thickness (IMT) as a surrogate marker for atherosclerosis. GCLM C-588T polymorphism was associated with average IMT (AveIMT) (r=0.090, p=0.0008), but the association between the other 3 polymorphisms and AveIMT did not reach the statistical significance. However, AveIMT was significantly greater as the total number of 4 concomitant "pro-oxidant alleles" in each subject was increased (r=0.108, p<0.0001). Furthermore, the number of "pro-oxidant alleles" was a risk factor for a high AveIMT independently of conventional risk factors (p=0.0003). In conclusion, accumulation of oxidative stress-associated alleles was associated with carotid atherosclerosis in type 2 diabetic patients.


Subject(s)
Atherosclerosis/genetics , Carotid Artery Diseases/genetics , Genetic Predisposition to Disease , Oxidative Stress/genetics , Polymorphism, Genetic , Alleles , Aryldialkylphosphatase/genetics , Atherosclerosis/complications , Atherosclerosis/pathology , Carotid Arteries/pathology , Carotid Artery Diseases/complications , Carotid Artery Diseases/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/enzymology , Female , Glutamate-Cysteine Ligase/genetics , Humans , Male , Peroxidase/genetics , Tunica Intima/pathology
2.
Transplant Proc ; 40(2): 438-40, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374094

ABSTRACT

BACKGROUND: We previously demonstrated that syngeneic pancreas transplantation has a potential to reverse diabetes even in a rat model of type 2 diabetes mellitus, namely Spontaneously Diabetic Torii (SDT; RT1a). The onset of diabetes was significantly delayed in the pancreas transplant recipients. We speculated that perfect diabetic control achieved by pancreas transplantation showed a beneficial effect on the native pancreata & recipients. MATERIALS AND METHODS: Twenty-five-week-old diabetic SDT rats were divided into 3 groups: untreated controls and syngeneic and allogeneic transplant recipients. We transplanted pancreaticoduodenal grafts from nondiabetic 10-week-old SDT rats and from 10-week-old allogeneic Dark Agouti (DA; RT1a) rats using daily administration of FK506. RESULTS: Untreated SDT rats showed disappearance of pancreatic and duodenal homeobox-1 (PDX-1) expression in the pancreas and a marked decrease in beta-cell mass. Among syngeneic and allogeneic pancreas transplant recipients, islet-like cell clusters were found in the native pancreata. The beta-cell mass at 40 weeks of age was significantly increased in the native pancreata of recipients compared with age-matched controls. Interestingly, we observed the reexpression of PDX-1 in the nuclei of islet-like cell clusters. CONCLUSIONS: Our results indicated the benefits of avoiding glucose toxicity by pancreas transplantation which induced PDX-1 expression in the native pancreata of type 2 diabetic recipients, resulting in regeneration of beta cells in the native pancreata.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 2/surgery , Insulin-Secreting Cells/physiology , Pancreas Transplantation , Animals , Body Weight , Homeodomain Proteins/genetics , Insulin-Secreting Cells/cytology , Male , Rats , Rats, Inbred Strains , Regeneration , Trans-Activators/genetics , Transplantation, Homologous , Transplantation, Isogeneic
3.
Diabetes Metab ; 44(3): 250-260, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29525225

ABSTRACT

AIMS: It is well-known that chronic exposure to large amounts of ligand leads to downregulation of its receptor. It is not known, however, whether a GLP-1R agonist downregulates its receptor. For this reason, our study examined whether GLP-1R expression is reduced after long-term exposure to dulaglutide (Dula) in non-diabetic and diabetic mice. METHODS: Seven-week-old male db/db and db/m mice were given either Dula (0.6mg/kg×2/week) or a control vehicle (CTL) for 17 weeks. Various metabolic parameters, such as glucose-stimulated insulin secretion (GSIS), insulin and TG content in islets, were evaluated after the intervention. ß-cell-related gene expression was also analyzed by real-time RT-PCR. RESULTS: In db/m mice, GLP-1R expression in ß-cells did not decrease, not even after long-term administration of Dula, compared with control mice, while GLP-1R expression in 24-week-old db/db mice treated with Dula was augmented, rather than downregulated, compared with 24-week-old CTL db/db mice. This was probably due to improved glycaemic control. In db/db mice treated with Dula, food intake and blood glucose levels were significantly decreased up to 24 weeks of age compared with CTL db/db mice, and their expression levels of various ß-cell-related genes, insulin content and GSIS were also enhanced. In contrast, oxidative and endoplasmic reticulum stress, inflammation, fibrosis and apoptosis were suppressed with Dula treatment. CONCLUSION: Dula exerts beneficial effects on glycaemic control and has long-lasting protective effects on pancreatic ß-cells. GLP-1R expression levels were not reduced at all in non-diabetic as well as diabetic mice despite long-term dulaglutide exposure.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptides/analogs & derivatives , Immunoglobulin Fc Fragments/pharmacology , Insulin-Secreting Cells/drug effects , Protective Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Blood Glucose/metabolism , Down-Regulation/drug effects , Glucagon-Like Peptides/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice
4.
J Clin Invest ; 100(7): 1840-6, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9312185

ABSTRACT

The insulin gene transcription factor PDX-1/IPF1/STF-1/ IDX-1 plays a key role in directing beta cell-specific gene expressions. Recently, impairment of PDX-1 expression or activity has been observed in beta cell-derived HIT cells cultured under high glucose concentrations, and this has been suggested as a possible cause of the decrease in insulin gene transcription. To investigate the pathophysiological significance of PDX-1 as a determinant of the rate of insulin gene transcription, we suppressed its expression in beta cell-derived MIN6 cells using an antisense oligodeoxynucleotide (ODN) and searched for possible changes in the beta cell-specific gene expression. Treatment of MIN6 cells with an 18-mer phosphorothioate ODN complementary to a sequence starting at the translation initiation codon of PDX-1 caused a potent, concentration-dependent reduction in PDX-1 expression; addition of 2 microM antisense ODN could reduce PDX-1 expression to 14+/-4% of the control. There was also a decrease in its DNA binding to the insulin gene A element. Despite such suppression of PDX-1, Northern blot analysis revealed no decrease in the amount of insulin mRNA in the MIN6 cells. Similarly, no changes were detected in the transcription of the glucokinase or islet amyloid polypeptide gene, for which PDX-1 was shown to function as a transcription factor. Thus, our findings dispute the physiological significance of PDX-1 in determining the rate of insulin gene transcription. This means that other components constituting the transcription-controlling machinery need to be evaluated in order to understand the molecular basis of impaired insulin biosynthesis such as that observed due to glucose toxicity.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins , Insulin/biosynthesis , Islets of Langerhans/metabolism , Trans-Activators/genetics , Binding Sites , Cell Line , Gene Expression , Glucose/pharmacology , Insulin/genetics , Islets of Langerhans/cytology , Oligonucleotides, Antisense/pharmacology , Protein Binding , RNA, Messenger/analysis , Regulatory Sequences, Nucleic Acid , Suppression, Genetic , Transcription, Genetic
5.
J Clin Invest ; 99(1): 144-50, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-9011569

ABSTRACT

Prolonged poor glycemic control in non-insulin-dependent diabetes mellitus patients often leads to a decline in insulin secretion from pancreatic beta cells, accompanied by a decrease in the insulin content of the cells. As a step toward elucidating the pathophysiological background of the so-called glucose toxicity to pancreatic beta cells, we induced glycation in HIT-T15 cells using a sugar with strong deoxidizing activity, D-ribose, and examined the effects on insulin gene transcription. The results of reporter gene analyses revealed that the insulin gene promoter is more sensitive to glycation than the control beta-actin gene promoter; approximately 50 and 80% of the insulin gene promoter activity was lost when the cells were kept for 3 d in the presence of 40 and 60 mM D-ribose, respectively. In agreement with this, decrease in the insulin mRNA and insulin content was observed in the glycation-induced cells. Also, gel mobility shift analyses using specific antiserum revealed decrease in the DNA-binding activity of an insulin gene transcription factor, PDX-1/IPF1/STF-1. These effects of D-ribose seemed almost irreversible but could be prevented by addition of 1 mM aminoguanidine or 10 mM N-acetylcysteine, thus suggesting that glycation and reactive oxygen species, generated through the glycation reaction, serve as mediators of the phenomena. These observations suggest that protein glycation in pancreatic beta cells, which occurs in vivo under chronic hyperglycemia, suppresses insulin gene transcription and thus can explain part of the beta cell glucose toxicity.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/metabolism , Homeodomain Proteins , Insulin/genetics , Insulin/metabolism , Promoter Regions, Genetic/drug effects , Ribose/pharmacology , Acetylcysteine/pharmacology , B-Lymphocytes , Blotting, Northern , Cells, Cultured , Cloning, Molecular , Genes, Reporter , Glucose/toxicity , Guanidines/pharmacology , Humans , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Ribose/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/physiology , Transcription, Genetic
6.
Mol Cell Biol ; 19(12): 8281-91, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10567553

ABSTRACT

Pax4 is a paired-domain (PD)-containing transcription factor which plays a crucial role in pancreatic beta/delta-cell development. In this study, we characterized the DNA-binding and transactivation properties of mouse Pax4. Repetitive rounds of PCR-based selection led to identification of the optimal DNA-binding sequences for the PD of Pax4. In agreement with the conservation of the optimal binding sequences among the Pax family transcription factors, Pax4 could bind to the potential binding sites for Pax6, another member of the Pax family also involved in endocrine pancreas development. The overexpression of Pax4 in HIT-T15 cells dose dependently inhibited the basal transcriptional activity as well as Pax6-induced activity. Detailed domain mapping analyses using GAL4-Pax4 chimeras revealed that the C-terminal region of Pax4 contains both activation and repression domains. The activation domain was active in the embryonic kidney-derived 293/293T cells and embryonal carcinoma-derived F9 cells, containing adenoviral E1A protein or E1A-like activity, respectively but was inactive or very weakly active in other cells including those of pancreatic beta- and alpha-cell origin. Indeed, the exogenous overexpression of type 13S E1A in heterologous cell types could convert the activation domain to an active one. On the other hand, the repression domain was active regardless of the cell type. When the repression domain was linked to the transactivation domain of a heterologous transcription factor, PDX-1, it could completely abolish the transactivation potential of PDX-1. These observations suggest a primary role of Pax4 as a transcriptional repressor whose function may involve the competitive inhibition of Pax6 function. The identification of the E1A-responsive transactivation domain, however, indicates that the function of Pax4 is subject to posttranslational regulation, providing further support for the complexity of mechanisms that regulate pancreas development.


Subject(s)
Adenovirus E1A Proteins/metabolism , Homeodomain Proteins/physiology , Pancreas/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Animals , Base Sequence , Binding Sites , Cell Line , Cell Line, Transformed , Cricetinae , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Islets of Langerhans/cytology , Mice , Molecular Sequence Data , Paired Box Transcription Factors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Response Elements , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
7.
Oncogene ; 19(46): 5298-302, 2000 Nov 02.
Article in English | MEDLINE | ID: mdl-11077447

ABSTRACT

The 14-3-3 sigma gene has been implicated in G2/M cell cycle arrest by p53. Frequent inactivation of the 14-3-3 sigma gene by hypermethylation of CpG islands has recently been reported in human breast carcinoma. The aim of this study was to examine the methylation status of CpG islands of the 14-3-3 sigma gene in hepatocellular carcinoma (HCC). The methylation status of the 14-3-3 sigma gene was evaluated in four normal liver tissues and 19 paired specimens of carcinoma and adjacent non-tumorous liver tissues using bisulfite-single strand conformation polymorphism (bisulfite-SSCP), a combination of sodium bisulfite modification and fluorescence-based polymerase chain reaction (PCR)-SSCP. The 14-3-3 sigma protein expression was examined by immunohistochemical staining. Hypermethylation of CpG islands of the 14-3-3 sigma gene was detected in 89% (17/19) of the HCC tissues but not in any of the four normal liver tissues. All of the 14 methylation-positive HCC samples analysed by immunohistochemistry showed loss of 14-3-3 sigma expression, while both of the methylation-negative HCC samples retained the expression, and a significant correlation was found between methylation and loss of expression. Lower levels of methylation were detected in adjacent non-tumorous liver tissues (6/16 in cirrhotic tissues and 1/3 in chronic hepatitis tissues), but the 14-3-3 sigma expression was retained in all of these tissues. In a methylation-positive HCC cell line, HLE, 5-aza-2'-deoxycytidine (5-aza-dC)-induced demethylation of CpG islands led to reactivation of gene expression, indicating that hypermethylation plays a causal role in inactivation of the 14-3-3 sigma gene in HCC. Hypermethylation and the resulting loss of expression of the 14-3-3 sigma gene corresponds to one of the most common abnormalities reported to date in HCC, suggesting their crucial role in the development and/or progression of HCC.


Subject(s)
Azacitidine/analogs & derivatives , Carcinoma, Hepatocellular/genetics , CpG Islands/genetics , Cytidine Triphosphate/analogs & derivatives , DNA Methylation , Gene Silencing , Liver Neoplasms/genetics , Tyrosine 3-Monooxygenase/genetics , 14-3-3 Proteins , Azacitidine/pharmacology , Base Sequence , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/pathology , Cytidine Triphosphate/pharmacology , Cytoplasm/chemistry , DNA Methylation/drug effects , DNA Mutational Analysis , Gene Silencing/drug effects , Humans , Immunohistochemistry , Liver/chemistry , Liver/metabolism , Liver/pathology , Liver Neoplasms/chemistry , Liver Neoplasms/pathology , Polymorphism, Single-Stranded Conformational , Sulfites , Tyrosine 3-Monooxygenase/analysis
8.
Diabetes ; 50 Suppl 1: S154-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272180

ABSTRACT

Inadequate beta-cell function is an essential component of all forms of diabetes. The most obvious problem is a failure to maintain sufficient beta-cell mass and function to cope with whatever insulin resistance is present. The most striking functional defect is a loss of acute glucose-induced insulin secretion (GIIS). This review discusses the ways in which beta-cells successfully adapt to increased demand and then decompensate as diabetes develops. Successful adaptation is achieved through increased beta-cell mass and increased insulin secretion. The hypothesis is explored that beta-cells exposed to the diabetic milieu lose their differentiation, which leads to loss of specialized functions such as GIIS. This concept has been strengthened by the finding of dedifferentiation of beta-cells in a rat model of partial pancreatectomy that includes a reduction of insulin gene expression, which may further contribute to decreased insulin production. Another finding was increased expression of c-Myc, which probably contributes to an increase in the expression of lactate dehydrogenase and the development of beta-cell hypertrophy. Arguments are developed that the beta-cell changes found in diabetes are better correlated with increased glucose levels than with non-esterified fatty acid levels, thus supporting the importance of glucose toxicity.


Subject(s)
Diabetes Mellitus/physiopathology , Islets of Langerhans/physiopathology , Adaptation, Physiological , Animals , Cell Division , Cell Size , Disease Progression , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Models, Biological
9.
Diabetes ; 45 Suppl 3: S81-3, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8674900

ABSTRACT

Under diabetic conditions, the Maillard reaction facilitates the production of reactive oxygen species, and the activity of antioxidant enzymes such as Cu,Zn-superoxide dismutase is decreased, resulting in a remarkable increase of oxidative stress. The oxidative stress attacks DNA, lipids, and proteins and is also thought to be involved in the pathogenesis of diabetic complications, including the progression of macroangiopathy. Proliferation of smooth muscle cells (SMCs) is known to be associated with progression of macroangiopathy and is modulated by several growth factors. At least three mitogens for SMCs, platelet-derived growth factor (PDGF), fibroblast growth factor, and heparin-binding epidermal growth factor-like growth factor (HB-EGF), are known to be produced by SMCs themselves and are considered to be the most potent growth factors in the progression of macroangiopathy as seen in diabetes. HB-EGF, but not PDGF, is regulated at the transcriptional level by 3-deoxyglucosone (3-DG), a major and highly reactive intermediate in the glycation reaction. The induction seems to be triggered by the increase of reactive oxygen species produced by 3-DG. Taken together, glycation reactions under diabetic conditions may be highly associated with the pathogenesis of diabetic macroangiography by enhancing the gene expression of HB-EGF.


Subject(s)
Diabetes Mellitus/metabolism , Diabetic Angiopathies/etiology , Glycation End Products, Advanced/metabolism , Oxidative Stress , Animals , Aorta , Arteriosclerosis/etiology , Arteriosclerosis/pathology , DNA Damage , Deoxyglucose/analogs & derivatives , Deoxyglucose/metabolism , Diabetic Angiopathies/metabolism , Epidermal Growth Factor/metabolism , Gene Expression , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins , Muscle, Smooth, Vascular/pathology , Oxidation-Reduction , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/genetics , Rats
10.
Diabetes ; 48(12): 2398-406, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10580429

ABSTRACT

Oxidative stress is produced under diabetic conditions and possibly causes various forms of tissue damage in patients with diabetes. The aim of this study was to examine the involvement of oxidative stress in the progression of pancreatic beta-cell dysfunction in type 2 diabetes and to evaluate the potential usefulness of antioxidants in the treatment of type 2 diabetes. We used diabetic C57BL/KsJ-db/db mice, in whom antioxidant treatment (N-acetyl-L-cysteine [NAC], vitamins C plus E, or both) was started at 6 weeks of age; its effects were evaluated at 10 and 16 weeks of age. According to an intraperitoneal glucose tolerance test, the treatment with NAC retained glucose-stimulated insulin secretion and moderately decreased blood glucose levels. Vitamins C and E were not effective when used alone but slightly effective when used in combination with NAC. No effect on insulin secretion was observed when the same set of antioxidants was given to nondiabetic control mice. Histologic analyses of the pancreases revealed that the beta-cell mass was significantly larger in the diabetic mice treated with the antioxidants than in the untreated mice. As a possible cause, the antioxidant treatment suppressed apoptosis in beta-cells without changing the rate of beta-cell proliferation, supporting the hypothesis that in chronic hyperglycemia, apoptosis induced by oxidative stress causes reduction of beta-cell mass. The antioxidant treatment also preserved the amounts of insulin content and insulin mRNA, making the extent of insulin degranulation less evident. Furthermore, expression of pancreatic and duodenal homeobox factor-1 (PDX-1), a beta-cell-specific transcription factor, was more clearly visible in the nuclei of islet cells after the antioxidant treatment. In conclusion, our observations indicate that antioxidant treatment can exert beneficial effects in diabetes, with preservation of in vivo beta-cell function. This finding suggests a potential usefulness of antioxidants for treating diabetes and provides further support for the implication of oxidative stress in beta-cell dysfunction in diabetes.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/physiopathology , Insulin Resistance , Islets of Langerhans/drug effects , Vitamin E/pharmacology , Animals , Blood Glucose/drug effects , Body Weight , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Female , Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
11.
Diabetes ; 44(7): 733-8, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7540572

ABSTRACT

Nitric oxide (NO) is believed to be an effector molecule that mediates interleukin (IL)-1 beta-induced destruction and dysfunction of pancreatic beta-cells. We have demonstrated that both exogenous NO and NO generated endogenously by IL-1 beta brought about apoptosis of isolated rat pancreatic islet cells as well as pancreatic beta-cell tumor-derived cell line HIT. This apoptosis was characterized by cleavage of DNA into nucleosomal fragments of 180-200 bp and morphologically by nuclear shrinkage, chromatic condensation, and apoptotic body formation. The IL-1 beta-induced internucleosomal DNA cleavage occurred in a time- and dose-dependent manner. Actinomycin D, cycloheximide, and nitric oxide synthase inhibitors inhibited the DNA cleavage, which was correlated with the amount of NO produced, indicating that NO produced by HIT cells themselves could mediate the apoptosis. Furthermore, in the presence of tumor necrosis factor (TNF)-alpha, large amounts of NO were produced by IL-1 beta and DNA cleavage occurred more noticeably, although TNF-alpha alone did not generate NO. Streptozotocin (STZ), a diabetogenic reagent containing a nitroso moiety, also released NO and induced internucleosomal DNA cleavage in HIT cells. These results suggest that NO-induced internucleosomal DNA cleavage is an important initial step in the destruction and dysfunction of pancreatic beta-cells induced by inflammatory stimulation or treatment with STZ.


Subject(s)
Amino Acid Oxidoreductases/biosynthesis , Apoptosis/drug effects , Gene Expression , Interleukin-1/pharmacology , Islets of Langerhans/drug effects , Nitric Oxide/pharmacology , Nitroprusside/pharmacology , Penicillamine/analogs & derivatives , Animals , Cell Survival/drug effects , Cricetinae , Cycloheximide/pharmacology , DNA/drug effects , DNA/isolation & purification , DNA/metabolism , Dactinomycin/pharmacology , Electrophoresis, Agar Gel , Humans , Insulinoma , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Kinetics , Microscopy, Electron, Scanning , Nitric Oxide/biosynthesis , Nitric Oxide Synthase , Nucleosomes/drug effects , Nucleosomes/metabolism , Pancreatic Neoplasms , Penicillamine/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , S-Nitroso-N-Acetylpenicillamine , Time Factors , Tumor Cells, Cultured
12.
Diabetes ; 45(11): 1478-88, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8866550

ABSTRACT

The glycolytic enzyme glucokinase plays a primary role in the glucose-responsive secretion of insulin, and defects of this enzyme can cause NIDDM. As a step toward understanding the molecular basis of glucokinase (GK) gene regulation, we assessed the structure and regulation of the human GK gene beta-cell-type promoter. The results of reporter gene analyses using HIT-T15 cells revealed that the gene promoter was comprised of multiple cis-acting elements, including two primarily important cis-motifs: a palindrome structure, hPal-1, and the insulin gene cis-motif A element-like hUPE3. While both elements were bound specifically by nuclear proteins, it was the homeodomain-containing transcription factor insulin promoter factor 1 (IPF1)/STF-1/PDX-1 that bound to the hUPE3 site: IPF1, when expressed in CHO-K1 cells, became bound to the hUPE3 site and activated transcription. An anti-IPF1 antiserum used in gel-mobility shift analysis supershifted the DNA protein complex formed with the hUPE3 probe and nuclear extracts from HIT-T15 cells, thus supporting the involvement of IPF1 in GK gene activation in HIT-T15 cells. In contrast to the insulin gene, however, neither the synergistic effect of the Pan1 expression on the IPF1-induced promoter activation nor the glucose responsiveness of the activity was observed for the GK gene promoter. These results revealed some conservative but unique features for the transcriptional regulation of the beta-cell-specific genes in humans. Being implicated in insulin and GK gene regulations as a common transcription factor, IPF1/STF-1/PDX-1 is likely to play an essential role in maintaining normal beta-cell functions.


Subject(s)
Glucokinase/genetics , Insulin/genetics , Islets of Langerhans/enzymology , Promoter Regions, Genetic , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Antibodies , Base Sequence , Binding Sites , CHO Cells , Cell Line , Cricetinae , Genes, Reporter , Glucokinase/biosynthesis , Homeodomain Proteins/metabolism , Humans , Luciferases/biosynthesis , Mesocricetus , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/immunology , Polymerase Chain Reaction , Restriction Mapping , Trans-Activators/analysis , Trans-Activators/chemistry , Transfection
14.
Free Radic Biol Med ; 22(6): 1007-17, 1997.
Article in English | MEDLINE | ID: mdl-9034240

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1) is a multifunctional polypeptide that is related to the progression of chronic pancreatitis. However, the mechanism of beta-cell damage by TGF-beta1 is unknown. Treatment with TGF-beta1 enhanced internucleosomal DNA cleavage caused by exogenous hydrogen peroxide in a hamster pancreatic beta-cell line (HIT). TGF-beta1 also induced protein oxidation, assessed by measuring carbonyl groups in proteins, and was involved in reactions that lead to lipid peroxidation. This eventually destructs membrane lipids and forms malondialdehyde. We have investigated its effects on two major antioxidative enzymes, catalase and glutathione peroxidase (GPx). TGF-beta1 suppressed mRNA expression as well as reduced the activities of catalase and GPx. The decrease in the catalase and GPx activities in TGF-beta1-treated cells resulted in an increase in intracellular peroxides as judged by flow cytometric analysis using a peroxide-sensitive dye, 2',7'-dichlorofluorescin diacetate. These data suggest that the augmented production of reactive oxygen species by TGF-beta1 through suppression of antioxidative enzymes may cause cellular damage and consequent apoptosis and induce pancreatitis or diabetes.


Subject(s)
Apoptosis , Catalase/antagonists & inhibitors , Glutathione Peroxidase/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/pharmacology , Adenoma, Islet Cell/metabolism , Adenoma, Islet Cell/pathology , Animals , Catalase/genetics , Cricetinae , DNA/metabolism , Flow Cytometry , Gene Expression , Glutathione Peroxidase/genetics , Hydrogen Peroxide/pharmacology , Oxidation-Reduction , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peroxides/analysis , RNA, Messenger/metabolism , Tumor Cells, Cultured
15.
FEBS Lett ; 461(1-2): 47-51, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10561494

ABSTRACT

A suppression subtractive hybridization technique was used to identify reactive oxygen species (ROS)-regulated genes in rat vascular smooth muscle cells. Three genes out of 89 clones, identified as fibronectin, p105 coactivator and ECA39, showed increased expression after treatment with H(2)O(2). The mRNA expressions of these three genes were induced in a time- and dose-dependent manner, independent of protein kinase C activation. Immunohistochemical staining showed that the p105 coactivator expression was markedly induced in the neointima of balloon-injured rat carotid arteries. These results suggest that ROS may play an important role in the development of atherosclerosis by regulating the gene expressions we identified in this study.


Subject(s)
Aorta/metabolism , Carotid Arteries/metabolism , Muscle, Smooth, Vascular/metabolism , Nucleic Acid Hybridization/methods , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Transaminases , Animals , Blotting, Northern , Cells, Cultured , Fibronectins/metabolism , Gene Expression Regulation , Gene Library , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Male , Muscle Proteins/metabolism , Proteins/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Time Factors
16.
Int J Oncol ; 16(3): 543-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10675487

ABSTRACT

Widespread or high-frequency microsatellite instability (MSI) due to the defective DNA mismatch repair (MMR) occurs in the majority of hereditary non-polyposis colorectal cancer and a subset of sporadic malignant tumors. The incidence of MSI and underlying DNA MMR defects have been well characterized in gastrointestinal carcinogenesis, but not in hepatocarcinogenesis. To address the issue, we analyzed 55 Japanese hepatocellular carcinomas using several indicators of DNA MMR defects, such as microsatellite analysis, loss of heterozygosity (LOH) and mutation analysis of MMR genes, methylation of hMLH1 promoter, and frameshift mutations of mononucleotide repeat sequences within possible target genes. Mutation of beta2-microglobulin gene, which is presumably involved in MSI-positive tumor cell escape from immune surveillance was also examined. Some of these analyses were also carried out in 9 human liver cancer cell lines. None of the 3 quasi-monomorphic mononucleotide markers sensitive for MSI, BAT26, BAT25, and BAT34C4 presented shortened unstable alleles in any of the carcinoma, cirrhosis, chronic hepatitis tissues, or cell lines. LOH at MMR genes was infrequent (4.4 approximately 7.1%), and no mutations were detected. Neither hMLH1 hypermethylation nor frameshift mutation in the target genes was detected. No mutations were found in beta2-microglobulin. Widespread MSI due to the defective DNA MMR appears to play little if any part in Japanese hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Microsatellite Repeats , Adult , Aged , Base Pair Mismatch , Carcinoma, Hepatocellular/virology , DNA Repair/genetics , Female , Hepatitis B virus/isolation & purification , Humans , Liver Neoplasms/virology , Loss of Heterozygosity , Male , Middle Aged
17.
Psychopharmacology (Berl) ; 102(3): 329-32, 1990.
Article in English | MEDLINE | ID: mdl-2174567

ABSTRACT

Mice were trained to avoid electric shock (0.6 mA) in a step-through type passive avoidance learning task, retention being measured 24 h after the training trial. Morphine 10 mg/kg administered 30 min before the test trial (pretest) facilitated memory retrieval, and the effect was completely antagonized by 1 mg/kg naloxone, a selective mu-opioid receptor antagonist. On the other hand, pretest administration of 0.01-10 mg/kg DTLET, a selective delta-opioid receptor agonist, did not produce the same effect as morphine. Nor-binaltorphimine, a kappa-opioid receptor antagonist, did not antagonize the effect of pretest morphine, at doses of 1 and 2 mg/kg. These results suggest that the facilitation of memory retrieval by pretest morphine is mediated through mu- but not delta- or kappa-opioid receptors.


Subject(s)
Benzeneacetamides , Memory/drug effects , Morphine/pharmacology , Receptors, Opioid/drug effects , Analgesics/pharmacology , Animals , Avoidance Learning/drug effects , Electroshock , Male , Mice , Mice, Inbred Strains , Naloxone/pharmacology , Oligopeptides/pharmacology , Pyrrolidines/pharmacology , Receptors, Opioid/physiology , Receptors, Opioid, delta , Receptors, Opioid, kappa , Receptors, Opioid, mu
18.
Psychopharmacology (Berl) ; 100(1): 27-30, 1990.
Article in English | MEDLINE | ID: mdl-2296624

ABSTRACT

Mice were trained in step-down and step-through type passive avoidance learning tasks and given retention tests. Pre-training administration of morphine impaired retention, the effect recovering completely after an additional injection of the same dose of morphine given 30 min before the retention test. Amnesia produced by scopolamine, cycloheximide and electroconvulsive shock was also reversed by pre-test morphine. Pre-test saline also reversed the morphine-induced memory impairment to some extent, indicating that the recovery may partially be due to the state dependent effect. Thus, it is demonstrated that pre-test morphine not only state dependently but also directly reversed memory impairment in mice.


Subject(s)
Memory/drug effects , Morphine/pharmacology , Amnesia/chemically induced , Animals , Avoidance Learning/drug effects , Cycloheximide/pharmacology , Electroshock , Male , Mice , Scopolamine/pharmacology
19.
Psychopharmacology (Berl) ; 112(1): 134-41, 1993.
Article in English | MEDLINE | ID: mdl-7871003

ABSTRACT

Mice were trained to avoid electric shocks by means of step-down type passive avoidance learning tasks, and memory retention was measured 24 h after the training session. Memory impairment (amnesia) was produced by administering either p-chloroamphetamine (PCA), a serotonin (5-HT) releaser or scopolamine (SCOP), a muscarinic cholinoceptor antagonist, 30 min prior to the training session. Benzomorphans, 5-HT2 antagonists and acetylcholinesterase (AChE) inhibitors were administered immediately after the training session. PCA- but not SCOP-induced amnesia was attenuated by the post-training administration of two benzomorphans, (+)N-allylnormetazocine ((+)SKF-10,047) and (+/- )pentazocine ((+/- )PTZ). Similarly, PCA-induced amnesia was reversed by the post-training administration of 5-HT2 antagonists, ritanserin (RIT) and mianserin (MIA), but SCOP-induced amnesia was not. However, the AChE inhibitors, tetrahydroaminoacridine (THA) and physostigmine (PHY) attenuated both PCA- and SCOP-induced amnesia when administered immediately after the training session. These results indicated that benzomorphans and 5-HT2 antagonists have antiamnestic effects in mice, as do AChE inhibitors. In addition, it is interesting that the patterns of ameliorating effect of benzomorphans were similar to those of 5-HT2 antagonists, which differ from those of AChE inhibitors.


Subject(s)
Avoidance Learning/drug effects , Benzomorphans/pharmacology , Cholinesterase Inhibitors/pharmacology , Serotonin Antagonists/pharmacology , Acetylcholine/metabolism , Amnesia/chemically induced , Amnesia/prevention & control , Animals , Electroshock , Male , Mice , Muscarinic Antagonists , Receptors, Muscarinic/metabolism , p-Chloroamphetamine/pharmacology
20.
J Biochem ; 119(3): 553-8, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8830053

ABSTRACT

Nitric oxide (NO) is synthesized from L-arginine by three isoforms of NO synthase (NOS). It is essential to suppress the function of the inducible isoform (macNOS) for amelioration of some inflammatory diseases in which the cytotoxic effect of NO is involved. S-Ethylsiothiourea (S-EIU) was reported to be a potent and specific inhibitor of macNOS. We also confirmed that it rather specifically inhibited the activity of the purified macNOS and the formation of nitrite by RAW264.7 cells compared to NG-monomethyl-L-arginine (L-NMA) and NG-nitro-L-arginine (L-NNA), the other isoforms being less effective. S-EIU suppressed the release of nitrite and lactate dehydrogenase from rat vascular smooth muscle cells treated with interleukin-1 beta and forskolin more potently than L-NMA or L-NNA. S-EIU also slightly suppressed internucleosomal DNA cleavage in pancreatic beta-cells induced by NO produced by macNOS. Intravenous administration of either S-EIU at 0.1 mg/kg/min or L-NMA at 1 mg/kg/min increased the blood pressure but decreased the heart rate in normal rabbits, while aminoguanidine at 1 mg/kg/min affected neither cardiovascular function. These inhibitors at these doses caused recovery of the blood pressure in lipopolysaccharide-treated rabbits that exhibited lowered blood pressure similar to that in the case of septic shock. Although S-EIU seemed not to be an adequate inhibitor for therapeutic use in vivo due to its side effects on cardiovascular functions, it is one of the most potent inhibitors of macNOS among reported inhibitors in vitro.


Subject(s)
Blood Pressure/drug effects , Enzyme Inhibitors/pharmacology , Heart Rate/drug effects , Isothiuronium/analogs & derivatives , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Brain/enzymology , Cells, Cultured , Citrulline/metabolism , Colforsin/pharmacology , DNA/metabolism , Dose-Response Relationship, Drug , Interleukin-1/pharmacology , Isothiuronium/pharmacology , L-Lactate Dehydrogenase/metabolism , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase/metabolism , Nitrites/metabolism , Nucleosomes/genetics , Rabbits , Rats , Shock, Septic/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL