Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 300
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Regul Homeost Agents ; 29(1): 51-62, 2015.
Article in English | MEDLINE | ID: mdl-25864741

ABSTRACT

It is important to determine the immunological properties for the maintenance of health. We chose the Shikoku Walking Pilgrimage to assess the proper biomarkers for the evaluation of immunological properties. We examined whether the Shikoku Walking Pilgrimage could have a positive effect on the mental and physical health of walking participants by using several biomarkers proposed by our laboratory. Twelve non-randomized healthy male volunteers including 3 twice attendees walked the Shikoku Walking Pilgrimage distance of 58.9 km over 3 days. Plasma, serum, urine, and saliva were collected from the volunteers during the pilgrimage and at 1 week before and after it. Immunological biomarkers, including lipid metabolism, oxidative stress, immune function, and catecholamines, were measured. Additionally, mood state scores, alertness, autonomic nervous system activity, and body motion levels during sleep were assessed. A significant decrease was observed in the subjective tension-anxiety levels and in the concentrations of serum low-density lipoprotein cholesterol, plasma hydroxyoctadecadienoic acid (HODE), and urine adrenaline during the pilgrimage as compared to the values of these parameters before the participants embarked on the pilgrimage. The serum levels of brain-derived neurotrophic factor (BDNF) were significantly increased 1 week after the pilgrimage relative to those assessed previously. No significant differences in subjective fatigue and the flicker perception threshold were observed. These results suggest that the Shikoku Walking Pilgrimage can exert a positive effect on mental and physical health as particularly shown in the reduction of tensionanxiety and oxidative stress without the accompaniment of fatigue. HODE correlated significantly with typical immunological marker natural killer cell activity and immunoglobulin G. This suggests that there are promising biomarkers such as HODE, NK activity, BDNF, LDL-c, and IgG for assessing the immunological properties.


Subject(s)
Biomarkers/analysis , Immune System/physiology , Walking/physiology , Walking/psychology , Adult , Affect/physiology , Anxiety/immunology , Anxiety/metabolism , Biomarkers/blood , Biomarkers/urine , Brain-Derived Neurotrophic Factor/blood , Cholesterol, LDL/blood , Epinephrine/urine , Fatigue/immunology , Fatty Acids, Unsaturated/blood , Humans , Immunoglobulin G/blood , Male , Middle Aged , Oxidative Stress
2.
J Clin Invest ; 87(5): 1867-71, 1991 May.
Article in English | MEDLINE | ID: mdl-2022753

ABSTRACT

We studied whether a novel vasoconstrictor peptide, endothelin-1 (ET-1), is synthesized by and released from human carcinoma cell lines, and whether ET-1 stimulates proliferation of these tumor cells. ET-1-like immunoreactivity was released from both HeLa and HEp-2 cells as a function of time. Reverse-phase HPLC of the conditioned media from HeLa cells revealed a major peak coeluting with standard ET-1. Northern blot analysis demonstrated the expression of mRNA for ET-1 precursor in both tumor cell lines. Both cell lines contained a single class of specific binding sites for ET-1. ET-1 dose-dependently induced increases in cytosolic free Ca2+ concentration in fura-2-loaded tumor cells, whose effect was completely abolished by chelating extracellular Ca2+ or by Ca(2+)-channel blocker. ET-1 stimulated proliferation of the quiescent cell lines in a dose-dependent manner, whose effect was inhibited by Ca(2+)-channel blocker. Polyclonal antibody for ET-1 inhibited proliferation of these cell lines, whereas nonimmune serum had no effect. These results demonstrate that ET-1 is synthesized by and released from human epithelial carcinoma cell lines, and that exogenous and endogenous ET-1 stimulates proliferation of the cells possibly through Ca2+ influx, suggesting its role as an autocrine/paracrine growth factor for certain tumor cells.


Subject(s)
Endothelins/pharmacology , Growth Substances/pharmacology , Calcium/metabolism , Cell Division/drug effects , Dose-Response Relationship, Drug , Endothelins/analysis , Endothelins/genetics , HeLa Cells , Humans , Tumor Cells, Cultured
3.
J Clin Invest ; 98(8): 1809-17, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8878432

ABSTRACT

Fabry disease is an X-linked disorder of glycosphingolipid metabolism caused by a deficiency of alpha-galactosidase A (alpha-Gal A). We identified a novel mutation of alpha-Gal A gene in a family with Fabry disease, which converted a tyrosine at codon 365 to a stop and resulted in a truncation of the carboxy (C) terminus by 65 amino acid (AA) residues. In a heterozygote of this family, although the mutant and normal alleles were equally transcribed in cultured fibroblasts, lymphocyte alpha-Gal A activity was approximately 30% of the normal control and severe clinical symptoms were apparent. COS-1 cells transfected with this mutant cDNA showed a complete loss of its enzymatic activity. Furthermore, those cotransfected with mutant and wildtype cDNAs showed a lower alpha-Gal A activity than those with wild type alone (approximately 30% of wild type alone), which suggested the dominant negative effect of this mutation and implied the importance of the C terminus for its activity. Thus, we generated mutant cDNAs with various deletion of the C terminus, and analyzed. Unexpectedly, alpha-Gal A activity was enhanced by up to sixfold compared with wild-type when from 2 to 10 AA residues were deleted. In contrast, deletion of 12 or more AA acid residues resulted in a complete loss of enzyme activity. Our data suggest that the C-terminal region of alpha-Gal A plays an important role in the regulation of its enzyme activity.


Subject(s)
Fabry Disease/genetics , Mutation , alpha-Galactosidase/genetics , Amino Acid Sequence , Cloning, Molecular , Fabry Disease/therapy , Female , Heterozygote , Humans , Middle Aged , Molecular Sequence Data , Pedigree , Polymerase Chain Reaction , RNA, Messenger/chemistry , Structure-Activity Relationship , X Chromosome , alpha-Galactosidase/chemistry , alpha-Galactosidase/metabolism
4.
Mol Cell Biol ; 14(9): 5748-55, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8065309

ABSTRACT

We used targeted homologous recombination to disrupt one c-myc gene copy in a diploid fibroblast cell line and found that a twofold reduction in Myc expression resulted in lower exponential growth rates and a lengthening of the G0-to-S-phase transition (M. Shichiri, K. D. Hanson and J. M. Sedivy, Cell Growth Differ. 4:93-104, 1993). Myc is a transcription factor, and the number of target genes whose regulation could result in differential growth rates may be very large. We have approached this problem by examining effects of reduced c-myc expression in three broad areas: (i) secretion of growth factors, (ii) expression of growth factor receptors, and (iii) intracellular signal transduction between Myc and components of the intrinsic cell cycle clock. We have found no evidence that differential medium conditioning can account for the growth phenotypes. Likewise, the expression of receptors for platelet-derived growth factor, epidermal growth factor, basic fibroblast growth factor, and insulin-like growth factor I was the same in diploid and heterozygous cells (platelet-derived growth factor, epidermal growth factor, fibroblast growth factor, and insulin-like growth factor are the sole growth factors required by these cells for growth in serum-free medium). In contrast, expression of cyclin E, cyclin A, and Rb phosphorylation were delayed when quiescent c-myc heterozygous cells were stimulated to enter the cell cycle. Expression of cyclin D1, cyclin D3, and Cdk2 was not affected. The timing of cyclin E induction was the earliest observable effect of reduced Myc expression. Our data indicate that Myc contributes to regulation of proliferation by a cell-autonomous mechanism that involves the modulation of cyclin E expression and, consequently, progression through the restriction point of the cell cycle.


Subject(s)
Cell Cycle , Genes, myc , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line , Cyclins/physiology , Gene Expression , In Vitro Techniques , Mutagenesis, Insertional , Phosphorylation , RNA, Messenger/genetics , Rats , Receptors, Growth Factor/metabolism , Retinoblastoma Protein/metabolism
5.
Oncogene ; 8(4): 899-907, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8455944

ABSTRACT

Analysis of how external proliferation signals impinge on the regulation of the cell cycle is ideally performed in cells that are capable of normal physiological withdrawal into the quiescent (G0) phase of the cell cycle as well as resumption of growth following appropriate stimuli. Targeted homologous recombination (gene targeting) provides an important new approach to determine the function of specific genes in these cellular processes. Current gene targeting methodology necessitates the use of immortal and stably diploid cell lines. This report investigates several rodent cell lines, by both genetic and physiological criteria, for use in gene targeting studies of the G0 to G1 transition. All murine cell lines examined were aneuploid. Some rat cell lines were euploid by chromosome number, but three specific genes, c-myc, c-raf-1 and Rb, were not always diploid. Only one cell line, an early-passage subclone of the Rat-1 cell line, was diploid for c-myc, c-raf-1 and Rb. An hprt- derivative of this cell line was isolated (designated TGR-1) and its karyotype was established by G-banding. TGR-1 cells were shown to withdraw into G0 upon serum starvation and to uniformly enter S phase after refeeding. Expression patterns of the c-myc, c-raf-1 and Rb genes and several properties of the gene products were found to be normal. The frequency of targeted homologous recombination of the c-myc and c-raf-1 loci was found to be within values observed with other cell lines. Thus, by both genetic and physiological criteria the TGR-1 cell line is a good model system for the analysis of the roles of c-myc, c-raf-1 and Rb in signal transduction, and will probably prove useful in studies involving other genes.


Subject(s)
Cell Cycle , Cell Line , Animals , Gene Expression , Genes, Retinoblastoma , Genes, myc , Hypoxanthine Phosphoribosyltransferase/genetics , In Situ Hybridization , In Vitro Techniques , Karyotyping , Models, Biological , Ploidies , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Proto-Oncogenes , Rats , Recombination, Genetic
6.
Biochim Biophys Acta ; 1170(2): 143-50, 1993 Oct 13.
Article in English | MEDLINE | ID: mdl-8399338

ABSTRACT

We previously demonstrated that acetylated human high-density lipoprotein (acetyl-HDL) was recognized by a scavenger receptor of rat sinusoidal liver cells (Murakami, M., Horiuchi, S., Takata, K. and Morino, Y. (1987) J. Biochem. (Tokyo) 101, 729-741). The present study describes the interaction of acetyl-HDL with rat peritoneal macrophages in vitro. Acetylation of HDL enhanced its cell-association by 2-fold and cellular degradation by > 25-fold. The cell-association of [125I]acetyl-HDL was effectively inhibited by unlabeled acetyl-HDL (> 85%), whereas the inhibition by HDL or acetylated human low-density lipoprotein (acetyl-LDL) was partial (60% and 50%, respectively). However, when both HDL and acetyl-LDL were present, the cell-association of [125I]acetyl-HDL was effectively inhibited by > 80%, a level identical or closely similar to that by acetyl-HDL. The cellular degradation of [125I]acetyl-HDL was effectively suppressed by acetyl-LDL whereas the effect of HDL was much weaker. These findings indicate that acetyl-HDL is endocytosed by both the HDL receptor and the scavenger receptor for acetyl-LDL in which the ligands bound to the latter might be subjected to lysosomal degradation.


Subject(s)
Carrier Proteins , Lipoproteins, HDL/metabolism , Macrophages/metabolism , RNA-Binding Proteins , Receptors, Lipoprotein/analysis , Acetylation , Animals , Cholesterol Esters/analysis , Iodine Radioisotopes , Lipoproteins, HDL/pharmacology , Lipoproteins, LDL/pharmacology , Lysosomes/metabolism , Male , Rats , Rats, Wistar , Receptors, LDL/analysis
7.
Biochim Biophys Acta ; 634(1): 1-10, 1981 Jan 14.
Article in English | MEDLINE | ID: mdl-6258642

ABSTRACT

Utilizing reflectance spectrophotometry, hemoperfusion, rate of oxygen consumption and redox level of mitochondrial cytochrome c (+c1) in livers in situ of anesthetized rats were measured. The transition to the anoxic state was induced by raising the pressure on the liver surface to more than the hepatic blood pressure by pressing with the tip of the optical guide of the reflectance spectrophotometer. During this transition, the average oxygen saturation of hemoglobin in the liver in situ decreased linearly with time until it became 10--20% of the total. This was followed by reduction of mitochondrial cytochrome c (+c1), which reached completion in 10--20 s. The measured O2 consumption rate remained constant until the percentage of oxyhemoglobin in situ decreased to a critical level. There was then a decrease in the rate of O2 consumption which was accompanied by a progressive reduction of cytochrome c (+c1). It was shown that amounts of hemoglobin and mitochondrial respiratory chain cytochromes in the liver in situ could be measured non-invasively and could provide important signals for vital cellular functions. The changes in hemoperfusion and rate of O2 consumption of the liver in situ following ethanol ingestion were also shown in rats, and are briefly discussed with respect to possible application of this method to study the pathophysiology of tissues.


Subject(s)
Cytochrome c Group/metabolism , Mitochondria, Liver/metabolism , Oxygen Consumption , Animals , Chemotherapy, Cancer, Regional Perfusion , Cytochromes c1/metabolism , Hemoglobins/metabolism , Kinetics , Male , Oxidation-Reduction , Rats , Spectrophotometry
8.
Biochim Biophys Acta ; 1391(2): 193-203, 1998 Mar 30.
Article in English | MEDLINE | ID: mdl-9555010

ABSTRACT

Acyl-coenzyme A:cholesterol acyltransferase (ACAT) is an enzyme catalyzing the intracellular formation of cholesteryl esters from free cholesterol and fatty acyl-CoA. In the present study, we cloned rat ACAT cDNA and determined its tissue distribution. Rat ACAT cDNA, having a coding region of 1635 bp with its deduced protein sequence of 545 amino acids and two typical motifs such as signature sequences and leucine heptad motif, showed 83, 92 and 90% identity with human, mouse, and hamster ACAT, respectively. Expression of rat ACAT cDNA in A293 cells and CHO cells resulted in a 3.0 to 3.5-fold increase in the enzyme activity. Among twelve tissues examined, ACAT activity was highest in adrenal followed by liver and intestine while that of aorta was extremely low. The mRNA level was also the highest in adrenal among four tissues examined. However, in contrast to its high ACAT activity, the liver mRNA level was extremely low (adrenal >> intestine > aorta >> liver). Consistent with mRNA levels, immunohistochemical analyses with a specific ACAT antibody detected significant ACAT signals in adrenal and intestine but a negligible signal in liver. These results indicate that adrenal most abundantly expresses ACAT in rat. Furthermore, rat liver showed a high ACAT activity but an extremely low ACAT mRNA and negligible immunohistochemical reactivity, suggesting the presence of a structurally different ACAT protein(s) in rat liver.


Subject(s)
Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cloning, Molecular , Cricetinae , DNA Primers/genetics , DNA, Complementary/genetics , Gene Expression , Humans , Immunohistochemistry , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Homology, Amino Acid , Tissue Distribution
9.
Biochim Biophys Acta ; 1301(1-2): 76-84, 1996 May 31.
Article in English | MEDLINE | ID: mdl-8652654

ABSTRACT

Many studies have shown that sterols can stimulate acyl-coenzyme A:cholesterol acyltransferase (ACAT) activity in cells. To elucidate this mechanism, effects of sterol-mediated induction on both the enzyme activity of ACAT and its mRNA levels were studied in human hepatoblastoma cell line, HepG2 cells. When HepG2 cells were loaded with cholesterol and 25-hydroxycholesterol, both the whole-cell ACAT activity and the microsomal ACAT activity were increased by 85.1% and 41.3%. In contrast, cholesterol depletion of HepG2 cells with compactin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, resulted in a decrease in both the whole-cell and the microsomal ACAT activity by 46.4% and 58.3%. Under identical conditions, RT-PCR and Northern blotting analyses revealed that neither cholesterol loading nor cholesterol depletion of HepG2 cells altered the amounts of ACAT mRNA. Moreover, these treatments had no effect on the enzymatic ACAT activity determined by the reconstituted assay in which HepG2 cell homogenate had been supplemented in vitro with a saturating level of exogenous cholesterol. These results indicate that cholesterol-induced up-regulation of ACAT activity in HepG2 cells does not occur at the level of transcription, but rather at a posttranscriptional level.


Subject(s)
Cholesterol/pharmacology , RNA, Messenger/analysis , Sterol O-Acyltransferase/drug effects , Base Sequence , Blotting, Northern , Hepatoblastoma/enzymology , Humans , Lipoproteins, LDL/metabolism , Molecular Sequence Data , Polymerase Chain Reaction , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Tumor Cells, Cultured
10.
Diabetes ; 28(4): 272-5, 1979 Apr.
Article in English | MEDLINE | ID: mdl-374171

ABSTRACT

Since it is important to elucidate the precise significance of pancreatic A-cell hypersecretion in the pathogenesis of diabetes mellitus, the change in the immunoreactive glucagon (IRG) response to 100 g oral glucose challenges was studied in diabetics whose blood glucose responses and plasma immunoreactive insulin concentrations (IRI) simulated those in normal subjects with the aid of the artificial beta cell system that we developed originally. In six nonobese adult-onset and four insulin-dependent diabetics whose blood glucose responses and plasma insulin concentrations after 100 g oral glucose load were made equivalent to those seen in normal subjects by the artificial beta cell, the glucagon release was similar to the response in normal subjects. In one insulin-dependent diabetic with high anti-insulin-binding capacity, the blood glucose response after an oral glucose challenge was not normalized by the artificial beta cell and the glucagon secretion was paradoxically increased. This fact suggested that the paradoxic rise in glucagon, seen in response to an oral glucose load in some diabetics, is secondary to insulin deficiency.


Subject(s)
Artificial Organs , Diabetes Mellitus/blood , Glucagon/blood , Islets of Langerhans/physiology , Adult , Aged , Blood Glucose/metabolism , Glucose Tolerance Test , Humans , Insulin/blood , Middle Aged
11.
Diabetes ; 29(9): 762-5, 1980 Sep.
Article in English | MEDLINE | ID: mdl-7002690

ABSTRACT

To elucidate the precise significance of pancreatic A-cell hypersecretion in the pathogenesis of diabetes mellitus, the change in the immunoreactive glucagon (IRG) response to intravenous arginine was studied in both nonobese, hypoinsulinemic non-insulin-dependent (NIDDM) and insulin-dependent diabetic (IDDM) subjects whose blood glucose responses and plasma immunoreactive insulin (IRI) simulated those of healthy subjects with the aid of the artificial beta-cell system that we originally developed. In both five NIDDM and five IDDM subjects, blood glucose responses and plasma IRI after arginine challenges were made equivalent to those seen in healthy subjects by infusing insulin in response to blood glucose, revealing that previously exaggerated IRG responses were made completely similar to the responses in healthy subjects. In summary, these results clearly demonstrate that the exaggerated response of A-cell secretion against arginine challenges in insulin-deficient diabetics is secondary to insulin lack, and the perfect normalization of its response is achieved only when both plasma insulin concentration and glycemic control simulate those of healthy subjects.


Subject(s)
Arginine , Artificial Organs , Diabetes Mellitus/blood , Glucagon/blood , Insulin/therapeutic use , Islets of Langerhans/metabolism , Adult , Blood Glucose/metabolism , Diabetes Mellitus/drug therapy , Humans , Kinetics
12.
Diabetes ; 24(11): 971-6, 1975 Nov.
Article in English | MEDLINE | ID: mdl-1237436

ABSTRACT

Alloxan-diabetic rats with fasting blood glucose levels above 300 mg./100 ml. were treated with intrajejunal administration of water-in-oil-in-water (W/O/W) insulin emulsions via an indwelling catheter at a dose of either 25 or 50 U./100 gm. body weight, three times daily for five to fourteen days. The course of diabetes was followed by determinations of glucose levels in blood and urine. During treatment a significant reduction in urinary glucose levels was observed in all rats studied. In two rats treated with 25 U./100 gm., fasting blood glucose levels did not change significantly. In four of five rats treated with 50 U./100 gm., W/O/W insulin emulsions significantly lessened hyperglycemia during treatment. Quantitative estimates suggested that the effectiveness of 50 U./100 gm. of intrajejunal W/O/W insulin emulsions was comparable to that after intramuscular regular insulin at doses between 1 and 2 U./100 gm. These results would indicate that diabetes can be controlled by enteral administration of insulin preparations.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Insulin/therapeutic use , Animals , Blood Glucose/metabolism , Catheterization , Circadian Rhythm , Dose-Response Relationship, Drug , Emulsions , Evaluation Studies as Topic , Glycosuria , Injections, Intramuscular , Insulin/administration & dosage , Jejunum , Male , Oils , Rats , Time Factors , Water
13.
Diabetes ; 33(12): 1200-2, 1984 Dec.
Article in English | MEDLINE | ID: mdl-6389235

ABSTRACT

We succeeded in miniaturizing a needle-type glucose monitoring system with characteristics suitable for application in a wearable, closed-loop control system. A wearable artificial endocrine pancreas (12 X 15 X 6 cm, 400 g) consisting of a sensor, a microcomputer system that calculates insulin and glucagon infusion rates, and two roller pumps was developed. Continuous glucose monitoring by a glucose sensor inserted in the subcutaneous tissue of the forearm or abdomen of healthy and diabetic volunteers revealed that glucose concentrations in subcutaneous tissue were 10% lower than, but were highly correlated with, blood glucose concentrations in the range of 49-388 mg/dl. Glycemic control was established in diabetic patients by intravenously infusing insulin in response to measured glucose concentrations on a moment-to-moment basis for a period of several days. By comparing the glycemic control obtained in each patient treated with multiple insulin injections or open-loop subcutaneous insulin infusion, the superiority of feedback control with the system was clearly demonstrated. During continuous glycemic regulation, day-to-day variations of insulin requirements were recognized in both basal insulin infusion and postprandial insulin infusion rates in response to identical meals and exercise. These data suggest the feasibility of long-term glycemic control in diabetic subjects with a wearable artificial endocrine pancreas, and indicate that to overcome changes in individual metabolic characteristics on a moment-to-moment basis, a closed-loop glycemic control system may be essential for ambulatory diabetic patients.


Subject(s)
Blood Glucose/analysis , Diabetes Mellitus, Type 1/therapy , Insulin Infusion Systems , Insulin/blood , Adult , Circadian Rhythm , Humans , Male , Methods , Microcomputers
14.
Diabetes ; 46(6): 929-36, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9166661

ABSTRACT

Insulin receptor substrate-1 (IRS-1) is one of the major substrates of insulin receptor tyrosine kinase and mediates various insulin signals downstream. In this study, we have examined the impact of three natural IRS-1 mutations identified in NIDDM patients (G971R, P170R, and M209T) on insulin signaling. G971R is located near src homology 2 protein binding sites, and P170R and M209T are located in the phosphotyrosine binding domain of IRS-1. 32D-IR cells, stably overexpressing human insulin receptor, were transfected with wild-type human IRS-1 cDNA (WT) or three mutant IRS-1 cDNAs and analyzed. All the cell lines expressing mutant IRS-1 showed a significant reduction in [3H]thymidine incorporation compared with WT. Upon insulin stimulation, cells expressing G971R showed a 39% decrease (P < 0.005) in phosphatidylinositol 3-kinase (PI 3-kinase) activity, a 43% decrease (P < 0.01) in binding of the 85-kDa regulatory subunit of PI 3-kinase, and a 22% decrease (P < 0.05) in mitogen-activated protein kinase activity compared with those expressing WT. Cells expressing P170R and M209T showed slight but significant decreases in PI 3-kinase activity (17 and 14%, respectively; both P < 0.05) and in binding of p85 (22 and 16%, respectively; both P < 0.05) and a greater decrease in mitogen-activated protein kinase activity (41 and 43%, respectively; both P < 0.005) compared with WT. After insulin stimulation, cells expressing P170R and M209T showed significant decreases in IRS-1 phosphorylation (37 and 42%, respectively; both P < 0.05) and in IRS-1 binding to the insulin receptor (48 and 53%, respectively; P < 0.01) compared with WT. G971R showed no changes in IRS-1 phosphorylation and in IRS-1 binding to the insulin receptor compared with WT. These data suggest that the impaired mitogenic response of P170R and M209T was mainly due to reduced binding to the insulin receptor, whereas the impaired response of G971R was mainly due to reduced association with PI 3-kinase p85.


Subject(s)
Gene Expression Regulation/genetics , Insulin/pharmacology , Mutation/genetics , Phosphoproteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , DNA/biosynthesis , DNA, Complementary/genetics , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Humans , Insulin Receptor Substrate Proteins , Mice , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Phosphorylation , Precipitin Tests , Rats , Thymidine/metabolism
15.
Diabetes ; 47(4): 621-31, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9568696

ABSTRACT

It has been proposed that mitochondrial oxidative phosphorylation in pancreatic beta-cells plays an important role in insulin secretion. To examine the impact of mitochondrial dysfunction on insulin secretion, we created a MIN6 cell line that depleted mitochondrial DNA (mtDNA) by treatment with ethidium bromide (EtBr), and studied the response of the cell line to various secretagogues. MIN6 cells cultured with 0.5 microg/ml EtBr for over 2 months (termed MIN6 deltamt cells) revealed a marked (>90%) decrease in mtDNA content and a lack of mRNAs encoded by mtDNA. MIN6 deltamt cells showed the defects of cytochrome c oxidase activity, glucose- and leucine-induced increase in cellular ATP content, and respiratory chain-driven ATP synthesis, suggesting that MIN6 deltamt cells lost oxidative phosphorylation activity due to the selective disruption of the subunits of respiratory chain enzymes encoded by mtDNA. MIN6 deltamt cells also showed a decrease in glucose utilization, suggesting the impairment of the glycolytic pathway as well. After stimulation with glucose and leucine, MIN6 deltamt cells showed no response in insulin secretion or intracellular free Ca2+ concentration ([Ca2+]i). On the other hand, arginine stimulated insulin secretion and an increase in [Ca2+]i in MIN6 deltamt cells as in MIN6 cells. Glibenclamide also stimulated insulin secretion and an increase in [Ca2+]i in both types of cells, but the responses of MIN6 deltamt cells were significantly lower than those of MIN6 cells. These results suggest the importance of ATP production in insulin secretion and an increase in [Ca2+]i, both induced by glucose and leucine. Moreover, mitochondrial function turns out to be not essential but important for the activation of sulfonylurea-induced insulin secretion.


Subject(s)
DNA, Mitochondrial/biosynthesis , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Leucine/pharmacology , Sulfonylurea Compounds/pharmacology , Adenosine Triphosphate/biosynthesis , Calcium/metabolism , Cell Respiration , Ethidium/pharmacology , Glucose/metabolism , Histocytochemistry , Humans , Insulin/biosynthesis , Insulin/genetics , Insulin Secretion , Intracellular Fluid/metabolism , Islets of Langerhans/cytology , Mitochondria/enzymology , Oxidation-Reduction , Phosphorylation , RNA, Messenger/biosynthesis , Tumor Cells, Cultured
16.
Diabetes ; 46(3): 354-62, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032089

ABSTRACT

Insulin receptor substrate 1 (IRS-1) is one of the major substrates of insulin receptor tyrosine kinase and mediates multiple insulin signals downstream. We have previously shown that the levels of IRS-1 mRNA varied in different tissues. To elucidate the molecular mechanisms of the tissue specific regulation of IRS-1, we have studied the cis-acting elements and transacting factors in CHO and HepG2 cells. Using the chloramphenicol acetyltransferase (CAT) assay with the various deletion mutants of the IRS-1 promoter-CAT fusion plasmids, several regions responsible for positive or negative regulation in each cell line were identified. A region from -1645 to -1585 bp, which regulated expression negatively in CHO cells and positively in HepG2 cells, was further analyzed. Within this region a fragment from -1645 to -1605 bp upregulated the IRS-1 promoter only in HepG2 cells, whereas a fragment from -1605 to -1585 bp downregulated only in CHO cells. In the gel mobility shift assay, several nuclear proteins that bind to these fragments were detected, and among them, two nuclear proteins that bind to a potential E box (nucleotide [nt] -1635 to -1630) and two nuclear proteins that bind to a potential C/EBP binding site (nt -1599 to -1591) were identified in HepG2 and CHO cells, respectively. CAT assays using promoters mutated at the E box or at the C/EBP binding site revealed that these sequences were responsible for cell-specific regulation of the IRS-1 gene. We therefore concluded that the two nuclear proteins that bind to the E box regulate IRS-1 gene expression positively in HepG2 cells and the two nuclear proteins that bind to the C/EBP binding site regulate it negatively in CHO cells.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/biosynthesis , Promoter Regions, Genetic , Animals , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins , CHO Cells , Chloramphenicol O-Acetyltransferase/biosynthesis , Cricetinae , Humans , Insulin Receptor Substrate Proteins , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides , Phosphoproteins/genetics , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Substrate Specificity , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured
17.
Diabetes ; 46(3): 463-72, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032104

ABSTRACT

Long-term incubation of proteins with glucose leads to advanced glycation end products (AGEs) with fluorescence and a brown color. We recently demonstrated immunologically the intracellular AGE accumulation in smooth muscle cell (SMC)-derived foam cells in advanced atherosclerotic lesions. To understand the mechanism of AGE accumulation in these foam cells, we have now characterized the interaction of AGE proteins with rabbit-cultured arterial SMCs. In experiments at 4 degrees C, 125I-labeled AGE-bovine serum albumin (AGE-BSA) showed a dose-dependent saturable binding to SMCs with an apparent dissociation constant (Kd) of 4.0 microg/ml. In experiments at 37 degrees C, AGE-BSA underwent receptor-mediated endocytosis and subsequent lysosomal degradation. The endocytic uptake of 125I-AGE-BSA was effectively inhibited by unlabeled AGE proteins such as AGE-BSA and AGE-hemoglobin, but not by acetylated LDL and oxidized LDL, well-known ligands for the macrophage scavenger receptor (MSR). Moreover, the binding of 125I-AGE-BSA to SMCs was affected neither by amphoterin, a ligand for one type of the AGE receptor, named RAGE, nor by 2-(2-furoyl)-4(5)-(2-furanyl)-1H-imidazole-hexanoic acid-BSA, a ligand for the other AGE receptors, p60 and p90. This indicates that the endocytic uptake of AGE proteins by SMCs is mediated by an AGE receptor distinct from MSR, RAGE, p60, and p90. To examine the functional role of this AGE receptor, the migratory effects of AGE-BSA on these SMCs were tested. Incubation with 1-50 microg/ml of AGE-BSA for 14 h resulted in significant dose-dependent cell migration. The AGE-BSA-induced SMC migration was chemotactic in nature and was significantly inhibited (approximately 80%) by an antibody against transforming growth factor-beta (TGF-beta), and the amount of TGF-beta secreted into the culture medium from SMC by AGE-BSA was sevenfold higher than that of control, indicating that TGF-beta is involved in the AGE-induced SMC chemotaxis. These data suggest that AGE may play a role in SMC migration in advanced atherosclerotic lesions.


Subject(s)
Chemotaxis , Foam Cells/physiology , Glycation End Products, Advanced/pharmacology , Lipoproteins, LDL/pharmacology , Muscle, Smooth, Vascular/physiology , Receptors, Immunologic/physiology , Serum Albumin, Bovine/pharmacology , Animals , Aorta, Thoracic , Biological Assay , Cell Division/drug effects , Cells, Cultured , DNA/biosynthesis , Endocytosis , Glycation End Products, Advanced/metabolism , Humans , Lipoproteins/blood , Lipoproteins/isolation & purification , Lysosomes/metabolism , Muscle, Smooth, Vascular/drug effects , Rabbits , Receptor for Advanced Glycation End Products , Serum Albumin, Bovine/metabolism , Substrate Specificity , Transforming Growth Factor beta/biosynthesis
18.
Mol Endocrinol ; 13(8): 1353-63, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10446908

ABSTRACT

Adrenomedullin is a novel vasodilatory peptide originally isolated from pheochromocytoma. Recently, we found that adrenomedullin acts as an autocrine/paracrine apoptosis survival factor for rat endothelial cells. In the present study, we show that adrenomedullin induces the expression of Max, a heterodimeric partner of c-Myc, which may contribute to its ability to rescue endothelial cells from apoptosis. Max is a basic-helix-loop-helix-leucine zipper protein that forms heterodimers with its alternative partners, Mad and Mxi-1, to behave as an antagonist for Myc-Max heterodimer through competition for common DNA targets. The expression of Max is reported to be constitutive and more stable than c-Myc, and serum induces immediate c-Myc stimulation followed by modest Max up-regulation. In quiescent rat endothelial cells, adrenomedullin stimulated the expression of Max without affecting c-Myc. Quantitation with real-time quantitative PCR detected on the ABI Prism 7700 Sequence Detection System revealed that adrenomedullin and calcitonin gene-related peptide (CGRP), as well as serum, up-regulated Max mRNA levels and that down-regulation of Max mRNA after serum deprivation was prevented by adrenomedullin. Neither adrenomedullin nor CGRP affected c-Myc expression. Transfection of a Max-expressing plasmid into endothelial cells rescued the apoptosis induced by serum deprivation. Neutralization with anti-adrenomedullin antiserum or blockade with a CGRP receptor antagonist, CGRP(8-37), reduced Max mRNA levels in growing endothelial cells and enhanced apoptosis after serum starvation. Introduction of an antisense oligodeoxynucleotide against Max mRNA using transferrin receptor-operated transfer led to inhibition of both adrenomedullin-induced up-regulation of Max transcripts and its cell survival effect, whereas random, sense, or missense oligonucleotides were without effect. The negative regulation of E-box-driven transcription by adrenomedullin was demonstrated by using preproendothelin-1 promoter containing c-Myc-Max binding consensus sequence; the promoter activity of preproendothelin-1 was reduced by cotransfecting Max- and Mad-expressing plasmids as well as addition of adrenomedullin and CGRP. The present results demonstrate that adrenomedullin antagonizes serum deprivation-induced endothelial apoptosis by up-regulation of the max gene in an autocrine/ paracrine manner.


Subject(s)
Apoptosis , Calcitonin Gene-Related Peptide/pharmacology , DNA-Binding Proteins/biosynthesis , Endothelium, Vascular/cytology , Peptides/pharmacology , Transcription Factors , Adrenomedullin , Animals , Aorta , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic-Leucine Zipper Transcription Factors , Blood , Cells, Cultured , DNA-Binding Proteins/genetics , Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Helix-Loop-Helix Motifs , Male , Oligodeoxyribonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Transfection
19.
Mol Endocrinol ; 12(2): 172-80, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9482660

ABSTRACT

Many vertebrate cells are resistant to apoptotic stimuli, whose variety and the mechanisms involved are not fully understood. Endothelin-1 is an endothelium-derived vasoactive peptide that mediates many physiological functions, such as vasoconstriction and cell proliferation. Deregulated expression of c-Myc induces apoptosis in serum-deprived fibroblasts. Using a panel of isogenic fibroblast cell lines with differential c-myc expression levels, we demonstrate that low doses of endothelin-1 protect fibroblasts against serum deprivation-induced apoptosis, which occurs through a c-Myc-dependent process. The endothelin-1-induced cell survival was mediated by the ET(A) receptor and was not linked to the ability of endothelin-1 to induce cell proliferation. The survival function of endothelin-1 was abrogated by inhibiting the mitogen-activated protein kinase pathway. These results demonstrate a hitherto unappreciated role of endothelin-1 as a potent survival factor for c-Myc-dependent apoptosis, a process mediated by the ET(A) receptor and the mitogen-activated protein kinase pathway.


Subject(s)
Apoptosis/drug effects , Endothelin-1/pharmacology , Proto-Oncogene Proteins c-myc/physiology , Animals , Apoptosis/genetics , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Culture Media, Serum-Free , Fibroblasts , Gene Expression Regulation/drug effects , Rats , Receptor, Endothelin A , Receptors, Endothelin/drug effects , Receptors, Endothelin/physiology
20.
Mol Endocrinol ; 9(10): 1367-79, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8544845

ABSTRACT

To evaluate the potential for regulation of the insulin receptor substrate IRS-1, we have cloned the mouse IRS-1 gene, identified its promoter, and analyzed promoter activity in the basal state and in response to stimulation. The 5'-region of the mouse IRS-1 gene lacks typical CAAT and TATA boxes but contains nine potential Sp1 binding sites consistent with a housekeeping gene. The 5'-region of the IRS-1 gene also has significant regions of homology with the promoters of the progesterone receptor gene, the insulin-like growth factor I receptor gene, and the androgen receptor gene. Multiple transcription start sites were identified 0.4-1.2 kilobases (kb) upstream from the start codon. Using a chloramphenicol acetyl transferase assay in Chinese hamster ovary (CHO) cells, basal promoter activity was present in the 3.2 kb 5'-flanking region of IRS-1 gene. Within this region, there were 184-base pair and 60-base pair negative regulatory elements at -3.2 kb and -1.6 kb surrounded by positive elements. By gel shift assay, a nuclear factor was identified in CHO cells which binds to -1606 and -1586 sequence in the negative regulatory element and appears to be distinct from C/EBP, CREB, and AP-1. In 3T3-F442A adipocytes dexamethasone treatment significantly decreased IRS-1 mRNA and IRS-1 protein. This was due to a decrease in the half-life of IRS-1 mRNA, with no change in IRS-1 promoter-chloramphenicol acetyl transferase activity. Insulin also decreased IRS-1 protein by approximately 60% within 9 h but did so without altering IRS-1 mRNA levels or chloramphenicol acetyl transferase activity. Thus, both insulin and dexamethasone down-regulate IRS-1 expression at the posttranscriptional level; with insulin this is probably due to an effect on protein half-life, whereas with dexamethasone the effect is due to a change in the half-life of IRS-1 mRNA.


Subject(s)
Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Animals , Base Sequence , CHO Cells , Cloning, Molecular , Cricetinae , Gene Expression Regulation , Gene Transfer Techniques , Insulin Receptor Substrate Proteins , Mice , Molecular Sequence Data , Phosphoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL