Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 14: 1167934, 2023.
Article in English | MEDLINE | ID: mdl-37251332

ABSTRACT

Hachimijiogan (HJG) has originally been used to ameliorate a variety of symptoms associated with low ambient temperatures. However, its pharmacological action in metabolic organs remains unclear. We hypothesized that HJG may modulate metabolic function and have a potential therapeutic application to metabolic diseases. To test this hypothesis, we investigated metabolic action of HJG in mice. Male C57BL/6J mice chronically administered with HJG showed a reduction in adipocyte size with increased transcription of beige adipocyte-related genes in subcutaneous white adipose tissue. HJG-mixed high-fat diet (HFD)-fed mice showed alleviation of HFD-induced weight gain, adipocyte hypertrophy, liver steatosis with a significant reduction in circulating leptin and Fibroblast growth factor 21 despite no changes in food intake or oxygen consumption. Feeding an HJG-mixed HFD following 4-weeks of HFD feeding, while a limited effect on body weight, improved insulin sensitivity with a reversal of decreased circulating adiponectin. In addition, HJG improved insulin sensitivity in the leptin-deficient mice without significant effects on body weight. Treatment with n-butanol soluble extracts of HJG potentiated transcription of Uncoupling protein 1 mediated by ß3-adrenergic agonism in 3T3L1 adipocytes. These findings provide evidence that HJG modulates adipocyte function and may exert preventive or therapeutic effects against obesity and insulin resistance.

2.
Pharmacol Ther ; 235: 108159, 2022 07.
Article in English | MEDLINE | ID: mdl-35183589

ABSTRACT

Bamboo is a medicinal plant, and has long been used as a traditional/folk medicine and a food preservative in Japan. Bamboo leaf contains many active ingredients with medicinal benefits. In particular, recent studies demonstrated that bamboo leaf extract and its constituents have great potential to prevent infectious, inflammatory, cardiovascular, metabolic, and neurological/neuropsychiatric diseases. In this review, we summarize the prophylactic and possible therapeutic effects of bamboo leaf extract and its constituent compounds against these disorders. The effects of the extract are explainable in part by the effects of some constituent compounds: p-coumaric acid, myricetin, orientin, stachyose, and vitexin. Moreover, coenzyme Q10, an anti-oxidative constituent, alleviates oxidative stress which underlies the common pathogenic mechanisms of the development of diabetic complications, atherosclerosis and periodontal disease. Some flavonoids contained in bamboo leaf, such as orientin and vitexin, have been reported to regulate gut microbiota responsible for maintaining whole-body functions, suggesting a possible interaction between bamboo leaf extract and probiotics. Thus, bamboo leaf is a valuable natural resource for the development of multiple pharmacotherapies.


Subject(s)
Atherosclerosis , Plant Extracts , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Humans , Oxidative Stress , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
3.
Phytomedicine ; 22(1): 16-22, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25636865

ABSTRACT

Sarcopenia is characterized by age-associated skeletal muscle atrophy and reduced muscle strength; currently, no pharmaceutical treatment is available. Go-sha-jinki-Gan (GJG) is a traditional Japanese herbal medicine that is used to alleviate various age-related symptoms, especially motor disorders. Here, we investigated the effect of GJG on aging-associated skeletal muscle atrophy by using senescence-accelerated mice (SAMP8). Immunohistochemical and western blotting analyses clearly showed that GJG significantly reduced the loss of skeletal muscle mass and ameliorated the increase in slow skeletal muscle fibers in SAMP8 mice compared to control mice. The expression levels of Akt and GSK-3ß, the phosphorylation of FoxO4, and the phosphorylations of AMPK and mitochondrial-related transcription factors such as PGC-1α were suppressed, while the expression of MuRF1 increased in SAMP8 mice, but approximated that in senescence-accelerated aging-resistant (SAMR1) mice after GJG treatment. We demonstrate for the first time that GJG has a therapeutic effect against sarcopenia.


Subject(s)
Aging , Drugs, Chinese Herbal/pharmacology , Muscle, Skeletal/drug effects , Sarcopenia/drug therapy , AMP-Activated Protein Kinases/metabolism , Animals , Cell Cycle Proteins , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin-Like Growth Factor I/metabolism , Male , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/metabolism , Muscle Strength/drug effects , Muscular Atrophy/drug therapy , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism
4.
Int J Oncol ; 38(1): 113-20, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21109932

ABSTRACT

The purpose of this study was to clarify the relationship between ovarian cancer peritoneal dissemination and indoleamine 2,3-dioxygenase (IDO) expression, and to explore the possibility of IDO-targeting molecular therapy for ovarian cancer. We transfected an IDO expression vector into the IDO-non-expressing human ovarian cancer cell line OMC-1, and established an IDO-expressing cell line (OMC-1/IDO) to examine the relationship between IDO expression and cancer cell growth in vitro and in vivo. IDO expression did not influence cancer cell growth and invasion in vitro, but promoted tumor growth and peritoneal dissemination in vivo. Immunostaining showed that IDO expression inhibited natural killer (NK) cell accumulation in tumors and promoted tumor angiogenesis. In addition, the oral administration of the IDO inhibitor 1-methly-tryptophan inhibited the growth of OMC-1/IDO-derived subcutaneous tumors in mice. These findings indicate that IDO promotes the peritoneal dissemination of ovarian cancer by inhibiting NK cell accumulation in tumors and promoting angiogenesis, supporting the applicability of IDO-targeting molecular therapy in ovarian cancer.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Killer Cells, Natural/immunology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/immunology , Peritoneal Neoplasms/immunology , Animals , Cell Line, Tumor , Disease Progression , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Transfection
5.
Mol Endocrinol ; 24(10): 1965-77, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20829391

ABSTRACT

Impairment of insulin and IGF-I signaling in the brain is one of the causes of dementia associated with diabetes mellitus and Alzheimer's disease. However, the precise pathological processes are largely unknown. In the present study, we found that SH2-containing inositol 5'-phosphatase 2 (SHIP2), a negative regulator of phosphatidylinositol 3,4,5-trisphosphate-mediated signals, is widely expressed in adult mouse brain. When a dominant-negative mutant of SHIP2 was expressed in cultured neurons, insulin signaling was augmented, indicating physiological significance of endogenous SHIP2 in neurons. Interestingly, SHIP2 mRNA and protein expression levels were significantly increased in the brain of type 2 diabetic db/db mice. To investigate the impact of increased expression of SHIP2 in the brain, we further employed transgenic mice overexpressing SHIP2 and found that increased amounts of SHIP2 induced the disruption of insulin/IGF-I signaling through Akt. Neuroprotective effects of insulin and IGF-I were significantly attenuated in cultured cerebellar granule neurons from SHIP2 transgenic mice. Consistently, terminal deoxynucleotide transferase-mediated dUTP nick end labeling assay demonstrated that the number of apoptosis-positive cells was increased in cerebral cortex of the transgenic mice at an elderly age. Furthermore, SHIP2 transgenic mice exhibited impaired memory performance in the Morris water maze, step-through passive avoidance, and novel-object-recognition tests. Importantly, inhibition of SHIP2 ameliorated the impairment of hippocampal synaptic plasticity and memory formation in db/db mice. These results suggest that SHIP2 is a potent negative regulator of insulin/IGF-I actions in the brain, and excess amounts of SHIP2 may be related, at least in part, to brain dysfunction in insulin resistance with type 2 diabetes.


Subject(s)
Brain/physiology , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Memory/physiology , Neuroprotective Agents/metabolism , Phosphoric Monoester Hydrolases/metabolism , Aging/physiology , Animals , Brain/cytology , Brain/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Enzyme Inhibitors/pharmacology , Inositol Polyphosphate 5-Phosphatases , Insulin Resistance/physiology , Memory/drug effects , Memory Disorders/physiopathology , Mice , Mice, Mutant Strains , Mice, Transgenic , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , Signal Transduction/physiology
6.
J Neurochem ; 110(3): 791-800, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19457071

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is the rate-limiting enzyme of the kynurenine pathway of tryptophan metabolism, ultimately leading to production of the excitotoxin quinolinic acid (QUIN) by monocytic cells. In the Tg2576 mouse model of Alzheimer's disease, systemic inflammation induced by lipopolysaccharide leads to an increase in IDO expression and QUIN production in microglia surrounding amyloid plaques. We examined whether the IDO over-expression in microglia could be mediated by brain proinflammatory cytokines induced during the peripheral inflammation using THP-1 cells and peripheral blood mononuclear cells (PBMC) as models for microglia. THP-1 cells pre-treated with 5-25 muM amyloid beta peptide (Abeta) (1-42) but not with Abeta (1-40) or Abeta (25-35) became an activated state as indicated by their morphological changes and enhanced adhesiveness. IDO expression was only slightly increased in the reactive cells but strongly enhanced following treatment with proinflammatory cytokine interferon-gamma (IFN-gamma) but not with interleukin-1beta, tumor necrosis factor-alpha, or interleukin-6 at 100 U/mL. The concomitant addition of Abeta (1-42) with IFN-gamma was totally ineffective, indicating that Abeta pre-treatment is prerequisite for a high IDO expression. The priming effect of Abeta (1-42) for the IDO induction was also observed for PBMC. These findings suggest that IFN-gamma induces IDO over-expression in the primed microglia surrounding amyloid plaques.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Inflammation Mediators/physiology , Interferon-gamma/physiology , Monocytes/enzymology , Peptide Fragments/physiology , Alzheimer Disease/enzymology , Animals , Cell Line, Tumor , Cells, Cultured , Enzyme Induction/physiology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Kynurenine/physiology , Macrophage Activation/physiology , Mice , Monocytes/pathology , Recombinant Proteins/pharmacology , Signal Transduction/physiology
7.
Endocrinology ; 149(2): 642-50, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18039790

ABSTRACT

SH2-containing inositol 5'-phosphatase 2 (SHIP2) is a 5'-lipid phosphatase hydrolyzing the phosphatidylinositol (PI) 3-kinase product PI(3,4,5)P(3) to PI(3,4)P(2) in the regulation of insulin signaling, and is shown to be increased in peripheral tissues of diabetic C57BL/KSJ-db/db mice. To clarify the impact of SHIP2 in the pathogenesis of insulin resistance with type 2 diabetes, we generated transgenic mice overexpressing SHIP2. The body weight of transgenic mice increased by 5.0% (P < 0.05) compared with control wild-type littermates on a normal chow diet, but not on a high-fat diet. Glucose tolerance and insulin sensitivity were mildly but significantly impaired in the transgenic mice only when maintained on the normal chow diet, as shown by 1.2-fold increase in glucose area under the curve over control levels at 9 months old. Insulin-induced phosphorylation of Akt was decreased in the SHIP2-overexpressing fat, skeletal muscle, and liver. In addition, the expression of hepatic mRNAs for glucose-6-phosphatase and phosphoenolpyruvate carboxykinase was increased, that for sterol regulatory element-binding protein 1 was unchanged, and that for glucokinase was decreased. Consistently, hepatic glycogen content was reduced in the 9-month-old transgenic mice. Structure and insulin content were histologically normal in the pancreatic islets of transgenic mice. These results indicate that increased abundance of SHIP2 in vivo contributes, at least in part, to the impairment of glucose metabolism and insulin sensitivity on a normal chow diet, possibly by attenuating peripheral insulin signaling and by altering hepatic gene expression for glucose homeostasis.


Subject(s)
Blood Glucose/metabolism , Insulin/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction/physiology , Adipocytes/cytology , Adipocytes/metabolism , Animals , Body Weight/physiology , Energy Metabolism/physiology , Female , Glycogen/metabolism , Homeostasis/physiology , Inositol Polyphosphate 5-Phosphatases , Ion Channels/genetics , Lipogenesis/physiology , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins/genetics , Pancreas/cytology , Pancreas/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , RNA, Messenger/metabolism , Transgenes/physiology , Uncoupling Protein 1 , Uncoupling Protein 2 , Uncoupling Protein 3
8.
Mol Pharmacol ; 71(3): 777-86, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17132684

ABSTRACT

The alpha7 nicotinic acetylcholine receptor subunit (CHRNA7) gene harbors a high degree of polymorphism. In this study, we found a novel variant (1267 G to A) in exon 10 of the CHRNA7 gene in a Japanese population. This variant results in glycine-to-serine substitution at position 423 (G423S) located in the large cytoplasmic loop of the protein. To clarify the possibility that the G423S mutation alters the pharmacological properties of alpha7 receptors, acetylcholine (ACh)-elicited current through alpha7-G423S mutant receptors expressed in Xenopus laevis oocytes was measured using the two-electrode voltage-clamp technique. We found that the current elicited by ACh (1 mM, 5 s) through alpha7-G423S receptors, but not through alpha7 receptors, was significantly decreased by treatment with a protein kinase C activator, phorbol-12-myristate-13-acetate (PMA, 10-30 nM). In addition, PMA (10 nM) selectively promoted a progressive decrease in alpha7-G423S current induced by repetitive application of ACh pulses (1 mM, 0.1 s, 0.17-0.33 Hz) compared with alpha7 current. PMA also enhanced the inactivation of alpha7-G423S mutant receptors induced by a prolonged application of choline (30 microM) without affecting alpha7 receptor responses. Western blot analysis showed that the treatment with PMA (30 nM) increased the serine phosphorylation level of the alpha7-G423S mutant receptors but not that of the wild-type receptors. These findings demonstrate that the G423S mutation promotes receptor desensitization by a protein kinase C-dependent mechanism. Thus, we provide the first evidence that a variant in the human CHRNA7 gene alters the function of alpha7 nicotinic receptors.


Subject(s)
Nicotinic Agonists/pharmacology , Protein Kinase C/physiology , Receptors, Nicotinic/chemistry , Adult , Animals , Female , Humans , Male , Middle Aged , Mutation , Phosphorylation , Polymorphism, Single Nucleotide , Receptors, Nicotinic/genetics , Receptors, Nicotinic/physiology , Structure-Activity Relationship , Tetradecanoylphorbol Acetate/pharmacology , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
9.
Diabetes ; 54(7): 1958-67, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15983195

ABSTRACT

We investigated the role of hepatic SH2-containing inositol 5'-phosphatase 2 (SHIP2) in glucose metabolism in mice. Adenoviral vectors encoding wild-type SHIP2 (WT-SHIP2) and a dominant-negative SHIP2 (DeltaIP-SHIP2) were injected via the tail vein into db/+m and db/db mice, respectively. Four days later, amounts of hepatic SHIP2 protein were increased by fivefold. Insulin-induced phosphorylation of Akt in liver was impaired in WT-SHIP2-expressing db/+m mice, whereas the reduced phosphorylation was restored in DeltaIP-SHIP2-expressing db/db mice. The abundance of mRNA for glucose-6-phosphatase (G6Pase) and PEPCK was increased, that for glucokinase (GK) was unchanged, and that for sterol regulatory element-binding protein 1 (SREBP)-1 was decreased in hepatic WT-SHIP2-overexpressing db/+m mice. The increased expression of mRNA for G6Pase and PEPCK was partly suppressed, that for GK was further enhanced, and that for SREBP1 was unaltered by the expression of DeltaIP-SHIP2 in db/db mice. The hepatic expression did not affect insulin signaling in skeletal muscle and fat tissue in both mice. After oral glucose intake, blood glucose levels and plasma insulin concentrations were elevated in WT-SHIP2-expressing db/+m mice, while elevated values were decreased by the expression of DeltaIP-SHIP2 in db/db mice. These results indicate that hepatic SHIP2 has an impact in vivo on the glucose metabolism in both physiological and diabetic states possibly by regulating hepatic gene expression.


Subject(s)
Glucose/metabolism , Insulin/pharmacology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Animals , CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins , Gene Transfer Techniques , Genetic Vectors , Glucokinase/genetics , Glucokinase/metabolism , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Humans , Leptin/deficiency , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , RNA, Messenger/genetics , Rats , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1 , Transcription Factors
10.
J Clin Endocrinol Metab ; 90(5): 2911-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15687335

ABSTRACT

Src homology 2-containing 5'-inositol phosphatase 2 (SHIP2) is known to be one of lipid phosphatases converting PI(3,4,5)P3 to PI(3,4)P2 in the negative regulation of insulin signaling with the fundamental impact on the state of insulin resistance. To clarify the possible involvement of SHIP2 in the pathogenesis of human type 2 diabetes, we examined the relation of human SHIP2 gene polymorphisms to type 2 diabetes in a Japanese population. We identified 10 polymorphisms including four missense mutations. Among them, single nucleotide polymorphism (SNP)3 (L632I) was located in the 5'-phosphatase catalytic region, and SNP5 (N982S) was adjacent to the phosphotyrosine binding domain binding consensus motif in the C terminus. SNP3 was found more frequently in control subjects than in type 2 diabetic patients, suggesting that this mutation might protect from insulin resistance. Transfection study showed that expression of SNP3-SHIP2 inhibited insulin-induced PI(3,4,5)P3 production and Akt2 phosphorylation less potently than expression of wild-type SHIP2 in CHO-IR cells. Insulin-induced tyrosine phosphorylation of SNP5-SHIP2 was decreased compared with that of wild-type SHIP2, resulting in increased Shc/Grb2 association and MAPK activation. These results indicate that the polymorphisms of SHIP2 are implicated, at least in part, in type 2 diabetes, possibly by affecting the metabolic and/or mitogenic insulin signaling in the Japanese population.


Subject(s)
Insulin/pharmacology , Phosphoric Monoester Hydrolases/genetics , Polymorphism, Genetic , Signal Transduction , Aged , Female , Haplotypes , Humans , Inositol Phosphates/biosynthesis , Linkage Disequilibrium , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt
11.
Mol Pharmacol ; 66(4): 1061-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15258256

ABSTRACT

Nicotinic acetylcholine receptors are key molecules in cholinergic transmission in the nervous system. Because of their structural complexity, only a limited number of subtype-specific agonists and antagonists are available to study nicotinic receptor functions. To overcome this limitation, we used voltageclamp recordings to examine the effects of several frog skin alkaloids on acetylcholine-elicited currents in Xenopus laevis oocytes expressing major types of neuronal nicotinic receptors (alpha4beta2, alpha7, alpha3beta2, alpha3beta4, and alpha4beta4). We found that the 5,8-disubstituted indolizidine (-)-235B' acted as a potent noncompetitive blocker of alpha4beta2 nicotinic receptors (IC50 = 74 nM). This effect was highly selective for alpha4beta2 receptors compared with alpha3beta2, alpha3beta4, and alpha4beta4 receptors. The inhibition of alpha4beta2 currents by (-)-235B' was more pronounced as the acetylcholine concentration increased (from 10 nM to 100 microM). Moreover, the blockade of alpha4beta2 currents by (-)-235B' was voltage-dependent (more pronounced at hyperpolarized potentials) and use-dependent, indicating that (-)-235B' behaves as an open-channel blocker of this receptor. Several other 5,8-disubstituted indolizidines (5-n-propyl-8-n-butylindolizidines), two 5,6,8-trisubstituted indolizidines ((-)-223A and (+)-6-epi-223A), and a 1,4-disubstituted quinolizidine ((+)-207I) were less potent than (-)-235B', and none showed selectivity for alpha4beta2 receptors. The quinolizidine (-)-1-epi-207I and the tricyclic (+)-205B had 8.7- and 5.4-fold higher sensitivity, respectively, for inhibition of the alpha7 nicotinic receptor than for inhibition of the alpha4beta2 receptor. These results show that frog alkaloids alter the function of nicotinic receptors in a subtype-selective manner, suggesting that an analysis of these alkaloids may aid in the development of selective drugs to alter nicotinic cholinergic functions.


Subject(s)
Alkaloids/pharmacology , Nicotinic Antagonists/pharmacology , Oocytes/drug effects , Receptors, Nicotinic/metabolism , Animals , Heterocyclic Compounds, 3-Ring/pharmacology , Indolizines/pharmacology , Oocytes/metabolism , Quinolizines/pharmacology , Receptors, Nicotinic/drug effects , Xenopus laevis
12.
FEBS Lett ; 554(3): 450-4, 2003 Nov 20.
Article in English | MEDLINE | ID: mdl-14623110

ABSTRACT

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is known to act as a lipid phosphatase hydrolyzing phosphatidylinositol (PI)(3,4,5)P(3) to PI(4,5)P(2). Since the PI3-kinase product, PI(3,4,5)P(3), is an important second messenger leading to the metabolic action of insulin, PTEN functions as a potent negative regulator of insulin signaling and its gene is one of the possible candidates involved in susceptibility to the development of type 2 (non-insulin-dependent) diabetes. In the present study, we investigated the polymorphisms of the PTEN gene in Japanese patients with type 2 diabetes and non-diabetic control subjects. We identified three mutations of the gene in the type 2 diabetes patients. Among these mutations, the frequency of the substitution of C with G at position -9 (-9C-->G) (SNP1), located in the untranslated region of exon 1, was significantly higher in type 2 diabetic patients than in control subjects. In addition, transfection of the PTEN gene with SNP1 resulted in a significantly higher expression level of PTEN protein compared with that of the wild-type PTEN gene in Cos1 and Rat1 cells. Furthermore, insulin-induced phosphorylation of Akt in HIRc cells was decreased more greatly by transfection of SNP1 PTEN gene than that of wild-type PTEN gene. These findings suggest that the change of C to G at position -9 of the PTEN gene is associated with the insulin resistance of type 2 diabetes due possibly to a potentiated hydrolysis of the PI3-kinase product.


Subject(s)
5' Untranslated Regions/genetics , Diabetes Mellitus, Type 2/genetics , Phosphoric Monoester Hydrolases/genetics , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins/genetics , Animals , COS Cells , Cell Line , DNA Primers/chemistry , DNA Primers/genetics , Exons/genetics , Female , Gene Frequency/genetics , Humans , Insulin/pharmacology , Introns/genetics , Japan , Male , Middle Aged , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/metabolism , Polymorphism, Single Nucleotide/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Transfection , Tumor Suppressor Proteins/metabolism
13.
Mol Endocrinol ; 16(10): 2371-81, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12351701

ABSTRACT

Lipid phosphatase SHIP2 [Src homology 2 (SH2)-containing inositol 5'-phosphatase 2] has been shown to be a physiologically critical negative regulator of insulin signaling. We investigated the molecular mechanism by which SHIP2 negatively regulates insulin-induced phosphorylation of Akt, a key downstream molecule of phosphatidylinositol 3-kinase important for the biological action of insulin. Overexpression of wild-type SHIP2 (WT-SHIP2) inhibited insulin-induced phosphorylation of Akt at both Thr(308) and Ser(473) in Rat1 fibroblasts expressing insulin receptors. The degree of inhibition was less in the cells expressing either a mutant SHIP2 with R47Q change (R/Q-SHIP2) in the SH2 domain, or a mutant SHIP2 with Y987F change (Y/F-SHIP2) in the C-terminal tyrosine phosphorylation site. However, on addition of a myristoylation signal, WT-SHIP2, R/Q-SHIP2, and Y/F-SHIP2 all efficiently inhibited insulin-induced Akt phosphorylation at both residues, whereas a 5'-phosphatase-defective mutant SHIP2 (deltaIP-SHIP2) with the myristoylation signal did not. Interestingly, the degree of inhibition of Akt phosphorylation by R/Q-SHIP2 and Y/F-SHIP2 is well correlated with the extent of their association with Shc. In addition, overexpression of WT-Shc increased the insulin-induced association of SHIP2 with Shc, whereas a decrease in the amount of Shc on expression of antisense Shc mRNA led to a reduction in the SHIP2-Shc association. Furthermore, the inhibitory effect on insulin-induced Akt phosphorylation by WT-SHIP2 was decreased in antisense-Shc cells. These results indicate that the membrane localization of SHIP2 with its 5'-phosphatase activity is required for negative regulation of insulin-induced Akt phosphorylation and that the localization is regulated, at least in part, by the association of SHIP2 with Shc in Rat1 fibroblasts.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Cell Membrane/metabolism , Insulin/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases , Receptor, Insulin/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antisense Elements (Genetics) , Cells, Cultured , Feedback, Physiological , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Insulin/pharmacology , Molecular Sequence Data , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Point Mutation , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Receptor, Insulin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tyrosine/metabolism , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...