Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Oncotarget ; 9(69): 33149-33159, 2018 Sep 04.
Article in English | MEDLINE | ID: mdl-30237858

ABSTRACT

BACKGROUND: There are no biomarkers to facilitate the identification of patients with ulcerative colitis (UC) who are at high risk for developing colorectal cancer (CRC). In our current study, we used rectal tissues from UC patients to identify aberrant DNA methylations and evaluated whether they could be used to identify UC patients with coexisting colorectal neoplasia. RESULTS: Using a training set, we identified 484 differentially methylated regions (DMRs) with absolute delta beta-values > 0.1 in rectal mucosa by using the ChAMP algorithm. Next, pathway enrichment analysis was performed using 484 DMRs to select coordinately methylated DMRs, resulting in the selection of 187 aberrant DMRs in rectal tissues from UC-CRC. Then, the Elastic Net classification algorithm was performed to narrow down optimal aberrant DMRs, and we finally selected 11 DMRs as biomarkers for identification of UC-CRC patients. The 11 chosen DMRs could discriminate UC patients with or without CRC in a training set (area under the curve, 0.96) and the validation set (area under the curve, 0.81). CONCLUSIONS: In conclusion, we identified 11 DMRs that could identify UC patients with CRC complications. Prospective studies should further confirm the validity of these biomarkers. METHODS: We performed genome-wide DNA methylation profiles in rectal mucosal tissues (n = 48) from 24 UC-CRC and 24 UC patients in a training set. Next, we performed comprehensive DNA methylation analysis using rectal mucosal tissues (n = 16) from 8 UC-CRC and 8 UC patients for validation.

2.
Int Immunopharmacol ; 50: 55-60, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28628771

ABSTRACT

S-1 is an oral agent containing tegafur (a prodrug of 5-fluorouracil) that is used to treat various cancers, but adverse effects are frequent. Two pilot clinical studies have suggested that lentinan (LNT; ß-1,3-glucan) may reduce the incidence of adverse effects caused by S-1 therapy. In this study, we established a murine model for assessment of gastrointestinal toxicity associated with S-1 and studied the effect of LNT. S-1 was administered orally to BALB/c mice at the effective dose (8.3mg/kg, as tegafur equivalent) once daily (5days per week) for 3weeks. Stool consistency and intestinal specimens were examined. We investigated the effect of combined intravenous administration of LNT at 0.1mg, which is an effective dose in murine tumor models. We also investigated the effect of a single administration of S-1. During long-term administration of S-1, some mice had loose stools and an increase in apoptotic bodies was observed in the ileal crypts. An increase in apoptotic bodies was also noted after a single administration of S-1 (15mg/kg). Prior or concomitant administration of LNT inhibited the increase in apoptotic bodies in both settings. Administration of LNT also increased the accumulation of CD11b+TIM-4+ cells in the ileum, while depletion of these cells by liposomal clodronate diminished the inhibitory effect of LNT on S-1 toxicity. Combined administration of LNT with S-1 led to a decrease in apoptotic bodies in the ileal crypts, possibly because LNT promoted phagocytosis of damaged cells by CD11b+TIM-4+ cells.


Subject(s)
Colonic Neoplasms/drug therapy , Extracellular Vesicles/drug effects , Ileum/drug effects , Lentinan/therapeutic use , Oxonic Acid/therapeutic use , Tegafur/therapeutic use , Administration, Intravenous , Animals , CD11b Antigen/metabolism , Cell Line, Tumor , Drug Combinations , Extracellular Vesicles/pathology , Humans , Ileum/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Models, Animal
3.
PLoS One ; 12(6): e0179955, 2017.
Article in English | MEDLINE | ID: mdl-28644884

ABSTRACT

BACKGROUND: Formation of 43S and 48S preinitiation complexes plays an important role in muscle protein synthesis. There is no muscle-wasting mouse model caused by a repressed 43S preinitiation complex assembly. OBJECTIVE: The aim of the present study was to develop a convenient mouse model of skeletal muscle wasting with repressed 43S preinitiation complex assembly. MATERIAL AND METHODS: A ligand-activatable PERK derivative Fv2E-PERK causes the phosphorylation of eukaryotic initiation factor 2α (eIF2α), which inhibits 43S preinitiation complex assembly. Thus, muscle atrophic phenotypes, intracellular signaling pathways, and intracellular free amino acid profiles were investigated in human skeletal muscle α-actin (HSA) promoter-driven Fv2E-PERK transgenic (Tg) mice. RESULTS: HSA-Fv2E-PERK Tg mice treated with the artificial dimerizer AP20187 phosphorylates eIF2α in skeletal muscles and leads to severe muscle atrophy within a few days of ligand injection. Muscle atrophy was accompanied by a counter regulatory activation of mTORC1 signaling. Moreover, intracellular free amino acid levels were distinctively altered in the skeletal muscles of HSA-Fv2E-PERK Tg mice. CONCLUSIONS: As a novel model of muscle wasting, HSA-Fv2E-PERK Tg mice provide a convenient tool for studying the pathogenesis of muscle loss and for assessing putative therapeutics.


Subject(s)
Disease Models, Animal , Mice, Transgenic , Muscle, Skeletal , Muscular Atrophy , Actins/genetics , Actins/metabolism , Amino Acids/metabolism , Animals , Homeostasis/physiology , Humans , Intracellular Space/metabolism , Ligands , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Multiprotein Complexes/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Tacrolimus/analogs & derivatives , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
4.
FASEB J ; 30(2): 798-812, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26487695

ABSTRACT

The eukaryotic translation initiation factor 2α (eIF2α) phosphorylation-dependent integrated stress response (ISR), a component of the unfolded protein response, has long been known to regulate intermediary metabolism, but the details are poorly worked out. We report that profiling of mRNAs of transgenic mice harboring a ligand-activated skeletal muscle-specific derivative of the eIF2α protein kinase R-like ER kinase revealed the expected up-regulation of genes involved in amino acid biosynthesis and transport but also uncovered the induced expression and secretion of a myokine, fibroblast growth factor 21 (FGF21), that stimulates energy consumption and prevents obesity. The link between the ISR and FGF21 expression was further reinforced by the identification of a small-molecule ISR activator that promoted Fgf21 expression in cell-based screens and by implication of the ISR-inducible activating transcription factor 4 in the process. Our findings establish that eIF2α phosphorylation regulates not only cell-autonomous proteostasis and amino acid metabolism, but also affects non-cell-autonomous metabolic regulation by induced expression of a potent myokine.


Subject(s)
Amino Acids/metabolism , Energy Metabolism/physiology , Eukaryotic Initiation Factor-2/metabolism , Fibroblast Growth Factors/biosynthesis , Gene Expression Regulation/physiology , Muscle, Skeletal/metabolism , Unfolded Protein Response/physiology , Amino Acids/genetics , Animals , Eukaryotic Initiation Factor-2/genetics , Fibroblast Growth Factors/genetics , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Phosphorylation/genetics
5.
Oncotarget ; 6(10): 8339-52, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25826076

ABSTRACT

Reduced expression of microRNA122 (miR122), a liver-specific microRNA, is frequent in hepatocellular carcinoma (HCC). However, its biological significances remain poorly understood. Because deregulated amino acid levels in cancers can affect their biological behavior, we determined the amino acid levels in miR122-silenced mouse liver tissues, in which intracellular arginine levels were significantly increased. The increased intracellular arginine levels were through upregulation of the solute carrier family 7 (SLC7A1), a transporter of arginine and a direct target of miR122. Arginine is the substrate for nitric oxide (NO) synthetase, and intracellular NO levels were increased in miR122-silenced HCC cells, with increased resistance to sorafenib, a multikinase inhibitor. Conversely, maintenance of the miR122-silenced HCC cells in arginine-depleted culture media, as well as overexpression of miR122 in miR122-low-expressing HCC cells, reversed these effects and rendered the cells more sensitive to sorafenib. Using a reporter knock-in construct, chemical compounds were screened, and Wee1 kinase inhibitor was identified as upregulators of miR122 transcription, which increased the sensitivity of the cells to sorafenib. These results provide an insight into sorafenib resistance in miR122-low HCC, and suggest that arginine depletion or a combination of sorafenib with the identified compound may provide promising approaches to managing this HCC subset.


Subject(s)
Arginine/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , MicroRNAs/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Apoptosis/physiology , Arginine/deficiency , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/biosynthesis , MicroRNAs/genetics , Niacinamide/pharmacology , Sorafenib
6.
J Am Soc Nephrol ; 25(9): 1954-65, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24652795

ABSTRACT

Vascular calcification (VC) is a life-threatening complication of CKD. Severe protein restriction causes a shortage of essential amino acids, and exacerbates VC in rats. Therefore, we investigated the effects of dietary l-lysine, the first-limiting amino acid of cereal grains, on VC. Male Sprague-Dawley rats at age 13 weeks were divided randomly into four groups: low-protein (LP) diet (group LP), LP diet+adenine (group Ade), LP diet+adenine+glycine (group Gly) as a control amino acid group, and LP diet+adenine+l-lysine·HCl (group Lys). At age 18 weeks, group LP had no VC, whereas groups Ade and Gly had comparable levels of severe VC. l-Lysine supplementation almost completely ameliorated VC. Physical parameters and serum creatinine, urea nitrogen, and phosphate did not differ among groups Ade, Gly, and Lys. Notably, serum calcium in group Lys was slightly but significantly higher than in groups Ade and Gly. Dietary l-lysine strongly suppressed plasma intact parathyroid hormone in adenine rats and supported a proper bone-vascular axis. The conserved orientation of the femoral apatite in group Lys also evidenced the bone-protective effects of l-lysine. Dietary l-lysine elevated plasma alanine, proline, arginine, and homoarginine but not lysine. Analyses in vitro demonstrated that alanine and proline inhibit apoptosis of cultured vascular smooth muscle cells, and that arginine and homoarginine attenuate mineral precipitations in a supersaturated calcium/phosphate solution. In conclusion, dietary supplementation of l-lysine ameliorated VC by modifying key pathways that exacerbate VC.


Subject(s)
Lysine/administration & dosage , Uremia/diet therapy , Vascular Calcification/prevention & control , Adenine/administration & dosage , Alanine/pharmacology , Animals , Apoptosis/drug effects , Arginine/pharmacology , Calcium/blood , Calcium/urine , Calcium Phosphates/metabolism , Cells, Cultured , Chemical Precipitation/drug effects , Creatinine/urine , Dietary Supplements , Homoarginine/pharmacology , Humans , Lysine/blood , Lysine/pharmacology , Male , Metabolic Networks and Pathways/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Osteoporosis/prevention & control , Proline/pharmacology , Rats , Rats, Sprague-Dawley , Solutions , Uremia/chemically induced , Uremia/complications , Vascular Calcification/etiology , Vascular Calcification/metabolism
7.
Amino Acids ; 46(5): 1275-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24553827

ABSTRACT

Taurine deficiency has been suggested to contribute to the pathogenesis and complications of advanced hepatic diseases. The molecular basis for a low level of taurine associated with hepatic failure is largely unknown. Using carbon tetrachloride (CCl4)-induced cirrhotic rat model, we found that the activity and expression of cysteine dioxygenase (CDO), a rate-limiting enzyme in taurine synthesis, were significantly decreased in the liver of these rats. To investigate the underlying mechanisms for the suppression, we examined the effects of pathological cytokines on CDO expression in human hepatoma HepG2 cells. Among the several cytokines, transforming growth factor-ß (TGF-ß), one of the key mediators of fibrogenesis, suppressed Cdo1 gene transcription through the MEK/ERK pathway. Finally, we further examined potential effects of branched-chain amino acids (BCAA) on CDO expression, as it has been reported that oral BCAA supplementation increased plasma taurine level in the patients with liver cirrhosis. BCAA, especially leucine, promoted Cdo1 gene transcription, and attenuated TGF-ß-mediated suppression of Cdo1 gene expression. These results indicate that the low plasma level of taurine in advanced hepatic disease is due to decreased hepatic CDO expression, which can be partly attributed to suppressive effect of TGF-ß on Cdo1 gene transcription. Furthermore, our observation that BCAA promotes Cdo1 expression suggests that BCAA may be therapeutically useful to improve hepatic taurine metabolism and further suppress dysfunctions associated with low level of taurine in hepatic diseases.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/metabolism , Liver Cirrhosis/enzymology , Taurine/biosynthesis , Transforming Growth Factor beta1/metabolism , Animals , Cysteine Dioxygenase/genetics , Down-Regulation , Hep G2 Cells , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Rats , Rats, Sprague-Dawley
8.
Biochem Biophys Res Commun ; 437(1): 127-33, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23806690

ABSTRACT

BACKGROUND & AIMS: Concentrations of the branched-chain amino acid (BCAA) in the serum of patients with liver cirrhosis correlate with their liver function. Oral administration of BCAA can ameliorate hypoalbuminemia and hepatic encephalopathy. In this study, we aim to clarify the role of BCAA in regulating the replication of the hepatitis C virus (HCV). METHODS: HCV sub-genomic replicon cells, genome-length replicon cells, and cells infected with cell culture-infectious HCV (HCVcc) were cultured in media supplemented with various concentrations of BCAA, followed by evaluation of the replicon or HCV abundance. RESULTS: BCAA was capable of suppressing the HCV replicon in a dose-dependent manner and the effect was independent of the mTOR pathway. Of the three BCAAs, valine was identified as being responsible for suppressing the HCV replicon. Surprisingly, an abundance of HJ3-5(YH/QL), an HCVcc, in Huh7 cells was augmented by BCAA supplementation. In contrast, BCAA suppressed an abundance of HJ3-5(wild), an HCVcc that cannot assemble virus particle in Huh7 cells. Internal ribosome entry site of HCV was shown to be a target of BCAA. Single-cycle virus production assays using Huh7-25 cells, which lacked CD81 expression, revealed that BCAA, especially valine, promoted infectious virus particle formation with minimal effect on virus secretion. Thus, BCAA was found to have two opposing effects on HCV production: suppression of the HCV genome RNA replication and promotion of infectious virus formation. CONCLUSIONS: BCAA accelerates HCV production through promotion of infectious virus formation in infected cells despite its suppressive effect on HCV genome replication.


Subject(s)
Hepacivirus/physiology , RNA, Viral/metabolism , Valine/pharmacology , Virion/metabolism , Virus Replication/drug effects , Cell Line, Tumor , Humans , Janus Kinases/metabolism , Replicon/genetics , Ribosomes/drug effects , Ribosomes/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Virion/drug effects
9.
Gastroenterology ; 141(1): 128-40, 140.e1-2, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21458454

ABSTRACT

BACKGROUND & AIMS: Patients with advanced chronic hepatitis C (CH-C) often are malnourished, but the effects of malnutrition on interferon (IFN) signaling and response to treatment have not been determined. We assessed the importance of the nutritional state of the liver on IFN signaling and treatment response. METHODS: We studied data from 168 patients with CH-C who were treated with the combination of pegylated-IFN and ribavirin. Plasma concentrations of amino acids were measured by mass spectrometry. Liver gene expression profiles were obtained from 91 patients. Huh-7 cells were used to evaluate the IFN signaling pathway, mammalian target of rapamycin complex 1 (mTORC1), and forkhead box O (FoxO). Antiviral signaling induced by branched-chain amino acids (BCAAs) was determined using the in vitro hepatitis C virus replication system. RESULTS: Multivariate logistic regression analysis showed that Fischer's ratio was associated significantly with nonresponders, independent of interleukin-28B polymorphisms or the histologic stage of the liver. Fischer's ratio was correlated inversely with the expression of BCAA transaminase 1, and was affected by hepatic mTORC1 signaling. IFN stimulation was impaired substantially in Huh-7 cells grown in medium that was low in amino acid concentration, through repressed mTORC1 signaling, and increased Socs3 expression, which was regulated by Foxo3a. BCAA could restore impaired IFN signaling and inhibit hepatitis C virus replication under conditions of malnutrition. CONCLUSIONS: Malnutrition impaired IFN signaling by inhibiting mTORC1 and activating Socs3 signaling through Foxo3a. Increasing BCAAs to up-regulate IFN signaling might be used as a new therapeutic approach for patients with advanced CH-C.


Subject(s)
Antiviral Agents/therapeutic use , Forkhead Transcription Factors/metabolism , Hepatitis C, Chronic/drug therapy , Interferon-alpha/therapeutic use , Liver/drug effects , Malnutrition/metabolism , Nutritional Status , Polyethylene Glycols/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Adult , Aged , Base Sequence , Cell Line, Tumor , Drug Therapy, Combination , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Genotype , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/growth & development , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/diagnosis , Hepatitis C, Chronic/metabolism , Humans , Interferon alpha-2 , Interferons , Interleukins/genetics , Interleukins/metabolism , Japan , Liver/metabolism , Liver/virology , Liver Cirrhosis/metabolism , Liver Cirrhosis/virology , Male , Malnutrition/virology , Mechanistic Target of Rapamycin Complex 1 , Middle Aged , Molecular Sequence Data , Multiprotein Complexes , Odds Ratio , Polymorphism, Genetic , Proteins/genetics , Proteins/metabolism , RNA Interference , RNA, Viral/blood , Recombinant Proteins , Regression Analysis , Ribavirin/therapeutic use , Severity of Illness Index , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , TOR Serine-Threonine Kinases/genetics , Transaminases/metabolism , Transfection , Treatment Outcome , Viral Load , Virus Replication/drug effects
10.
Autophagy ; 7(7): 727-36, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21471734

ABSTRACT

Both anabolism and catabolism of the amino acids released by starvation-induced autophagy are essential for cell survival, but their actual metabolic contributions in adult animals are poorly understood. Herein, we report that, in mice, liver autophagy makes a significant contribution to the maintenance of blood glucose by converting amino acids to glucose via gluconeogenesis. Under a synchronous fasting-initiation regimen, autophagy was induced concomitantly with a fall in plasma insulin in the presence of stable glucagon levels, resulting in a robust amino acid release. In liver-specific autophagy (Atg7)-deficient mice, no amino acid release occurred and blood glucose levels continued to decrease in contrast to those of wild-type mice. Administration of serine (30 mg/animal) exerted a comparable effect, raising the blood glucose levels in both control wild-type and mutant mice under starvation. Thus, the absence of the amino acids that were released by autophagic proteolysis is a major reason for a decrease in blood glucose. Autophagic amino acid release in control wild-type livers was significantly suppressed by the prior administration of glucose, which elicited a prompt increase in plasma insulin levels. This indicates that insulin plays a dominant role over glucagon in controlling liver autophagy. These results are the first to show that liver-specific autophagy plays a role in blood glucose regulation.


Subject(s)
Amino Acids/blood , Autophagy , Blood Glucose/metabolism , Liver/cytology , Liver/metabolism , Animals , Fasting/blood , Fatty Acids/blood , Glucagon/blood , Gluconeogenesis , Insulin/blood , Liver/ultrastructure , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Muscle, Skeletal/metabolism , Starvation , Triglycerides/blood , Vacuoles/metabolism , Vacuoles/ultrastructure
11.
Cell Metab ; 13(2): 170-82, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21284984

ABSTRACT

Maintenance of skeletal muscle mass relies on the dynamic balance between anabolic and catabolic processes and is important for motility, systemic energy homeostasis, and viability. We identified direct target genes of the glucocorticoid receptor (GR) in skeletal muscle, i.e., REDD1 and KLF15. As well as REDD1, KLF15 inhibits mTOR activity, but via a distinct mechanism involving BCAT2 gene activation. Moreover, KLF15 upregulates the expression of the E3 ubiquitin ligases atrogin-1 and MuRF1 genes and negatively modulates myofiber size. Thus, GR is a liaison involving a variety of downstream molecular cascades toward muscle atrophy. Notably, mTOR activation inhibits GR transcription function and efficiently counteracts the catabolic processes provoked by glucocorticoids. This mutually exclusive crosstalk between GR and mTOR, a highly coordinated interaction between the catabolic hormone signal and the anabolic machinery, may be a rational mechanism for fine-tuning of muscle volume and a potential therapeutic target for muscle wasting.


Subject(s)
Muscle, Skeletal/metabolism , Receptors, Glucocorticoid/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Kruppel-Like Transcription Factors/metabolism , Mice , Muscle Proteins/metabolism , Protein Binding , Rats , Receptors, Glucocorticoid/genetics , Repressor Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Transcription Factors , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism
12.
J Biol Chem ; 285(26): 20109-16, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20427287

ABSTRACT

Mammalian target of rapamycin (mTOR) is a member of the phosphatidylinositol 3-kinase-related kinase (PIKK) family and is a major regulator of translation, cell growth, and autophagy. mTOR exists in two distinct complexes, mTORC1 and mTORC2, that differ in their subunit composition. In this study, we identified KIAA0406 as a novel mTOR-interacting protein. Because it has sequence homology with Schizosaccharomyces pombe Tti1, we named it mammalian Tti1. Tti1 constitutively interacts with mTOR in both mTORC1 and mTORC2. Knockdown of Tti1 suppresses phosphorylation of both mTORC1 substrates (S6K1 and 4E-BP1) and an mTORC2 substrate (Akt) and also induces autophagy. S. pombe Tti1 binds to Tel2, a protein whose mammalian homolog was recently reported to regulate the stability of PIKKs. We confirmed that Tti1 binds to Tel2 also in mammalian cells, and Tti1 interacts with and stabilizes all six members of the PIKK family of proteins (mTOR, ATM, ATR, DNA-PKcs, SMG-1, and TRRAP). Furthermore, using immunoprecipitation and size-exclusion chromatography analyses, we found that knockdown of either Tti1 or Tel2 causes disassembly of mTORC1 and mTORC2. These results indicate that Tti1 and Tel2 are important not only for mTOR stability but also for assembly of the mTOR complexes to maintain their activities.


Subject(s)
Carrier Proteins/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Carrier Proteins/genetics , Cell Line , Chromatography, Gel , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Kinases/metabolism , Proteins , Proto-Oncogene Proteins c-ets/genetics , RNA Interference , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Transfection
13.
Cell Tissue Res ; 336(2): 203-12, 2009 May.
Article in English | MEDLINE | ID: mdl-19290545

ABSTRACT

Amino acids have various physiological activities that influence processes such as intestinal regeneration, EGF secretion, protein synthesis, and cell growth. Salivary glands are exposed to nutrients that influence their proliferation and regeneration. Glycine is included in saliva in large quantities and reportedly has important roles in antibacterial activities and the inhibition of tumor growth and as a precursor of nucleotide synthesis in cell proliferation. We have investigated the effects of glycine on the proliferation and differentiation of salivary glands by using mouse salivary-gland-derived progenitor (mSGP) cells. In cultures of mSGP cells, cell proliferation is suppressed in the presence of glycine, whereas it is promoted by its removal. Glycine promotes three-dimensional formations of mSGP cells, which are negative for immature markers and positive for differentiation markers. In cell-cycle analysis, cell-cycle progression is delayed at the S-phase by glycine supplementation. Glycine also suppresses the phosphorylation of p42/p44MAPK. These results suggest that glycine suppresses the proliferation and promotes the differentiation of mSGP cells, and that it has inhibitory effects on growth factor signaling and cell-cycle progression. Glycine might therefore be a physiological activator that regulates the proliferation and differentiation of salivary glands.


Subject(s)
Cell Differentiation/drug effects , Glycine/pharmacology , Salivary Glands/cytology , Stem Cells/cytology , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Mice , S Phase/drug effects , Time Factors
14.
Mol Biol Cell ; 20(7): 1981-91, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19211835

ABSTRACT

Autophagy is an intracellular degradation system, by which cytoplasmic contents are degraded in lysosomes. Autophagy is dynamically induced by nutrient depletion to provide necessary amino acids within cells, thus helping them adapt to starvation. Although it has been suggested that mTOR is a major negative regulator of autophagy, how it controls autophagy has not yet been determined. Here, we report a novel mammalian autophagy factor, Atg13, which forms a stable approximately 3-MDa protein complex with ULK1 and FIP200. Atg13 localizes on the autophagic isolation membrane and is essential for autophagosome formation. In contrast to yeast counterparts, formation of the ULK1-Atg13-FIP200 complex is not altered by nutrient conditions. Importantly, mTORC1 is incorporated into the ULK1-Atg13-FIP200 complex through ULK1 in a nutrient-dependent manner and mTOR phosphorylates ULK1 and Atg13. ULK1 is dephosphorylated by rapamycin treatment or starvation. These data suggest that mTORC1 suppresses autophagy through direct regulation of the approximately 3-MDa ULK1-Atg13-FIP200 complex.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amino Acids/pharmacology , Autophagy/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Amino Acids/deficiency , Animals , Autophagy-Related Protein-1 Homolog , Autophagy-Related Proteins , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/ultrastructure , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Molecular Weight , Multiprotein Complexes , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinases/metabolism , Protein Transport/drug effects , Proteins , TOR Serine-Threonine Kinases
15.
Int J Biomed Sci ; 5(3): 293-301, 2009 Sep.
Article in English | MEDLINE | ID: mdl-23675150

ABSTRACT

Albumin (Alb) is mixture of reduced and oxidized forms. It is physiologically significant to determine Alb(red)%, which is the proportion of reduced Alb in the sum of Alb. However, reduced Alb in both blood and plasma samples is easily converted to oxidized Alb. Accordingly, the stabilization of Alb in samples is necessary to determine precise Alb(red)% values. Alb stabilization in blood or plasma was achieved by pH control and buffer dilution. At least a 50-fold dilution with 50 mmol/l phosphate buffer (pH 6.0) was required for human plasma. For human blood, a 10-fold dilution with 0.5 mol/l sodium citrate buffer (pH 4.3) was required. To measure Alb(red)%, treated samples were applied to HPLC or LC-ESI-TOFMS. We also developed a "pre-incubation method", to accelerate the oxidative reaction in plasma by heating at 37°C. Alb(red)% values were maintained around the initial value for 48 h after stabilizing human plasma and 72 h after stabilizing human blood. Accelerating the oxidative reaction in plasma produced large differences in the Alb(red)% values between normal and model disease samples. Precise Alb(red)% values were routinely obtained under the stabilization control. Additionally, pre-incubation of the plasma before measurement is useful to enhance the difference between normal and disease samples.

16.
J Biol Chem ; 282(28): 20329-39, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17517883

ABSTRACT

The proline-rich Akt substrate of 40 kilodaltons (PRAS40) was identified as a raptor-binding protein that is phosphorylated directly by mammalian target of rapamycin (mTOR) complex 1 (mTORC1) but not mTORC2 in vitro, predominantly at PRAS40 (Ser(183)). The binding of S6K1 and 4E-BP1 to raptor requires a TOR signaling (TOS) motif, which contains an essential Phe followed by four alternating acidic and small hydrophobic amino acids. PRAS40 binding to raptor was severely inhibited by mutation of PRAS40 (Phe(129) to Ala). Immediately carboxyl-terminal to Phe(129) are two small hydrophobic amino acid followed by two acidic residues. PRAS40 binding to raptor was also abolished by mutation of the major mTORC1 phosphorylation site, Ser(183), to Asp. PRAS40 (Ser(183)) was phosphorylated in intact cells; this phosphorylation was inhibited by rapamycin, by 2-deoxyglucose, and by overexpression of the tuberous sclerosis complex heterodimer. PRAS40 (Ser(183)) phosphorylation was also inhibited reversibly by withdrawal of all or of only the branched chain amino acids; this inhibition was reversed by overexpression of the Rheb GTPase. Overexpressed PRAS40 suppressed the phosphorylation of S6K1 and 4E-BP1 at their rapamycin-sensitive phosphorylation sites, and reciprocally, overexpression of S6K1 or 4E-BP1 suppressed phosphorylation of PRAS40 (Ser(183)) and its binding to raptor. RNA interference-induced depletion of PRAS40 enhanced the amino acid-stimulated phosphorylation of both S6K1 and 4E-BP1. These results establish PRAS40 as a physiological mTORC1 substrate that contains a variant TOS motif. Moreover, they indicate that the ability of raptor to bind endogenous substrates is limiting for the activity of mTORC1 in vivo and is therefore a potential locus of regulation.


Subject(s)
Phosphoproteins/metabolism , Protein Processing, Post-Translational/physiology , Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Substitution , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Gene Expression , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes , Mutation, Missense , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphoproteins/genetics , Phosphorylation , Protein Binding/physiology , Proteins/genetics , RNA Interference , Ras Homolog Enriched in Brain Protein , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/genetics
17.
FEBS J ; 273(14): 3346-57, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16857017

ABSTRACT

Human serum albumin (HSA) exists in both reduced and oxidized forms, and the percentage of oxidized albumin increases in several diseases. However, little is known regarding the pathophysiological significance of oxidation due to poor characterization of the precise structural and functional properties of oxidized HSA. Here, we characterize both the structural and functional differences between reduced and oxidized HSA. Using LC-ESI-TOFMS and FTMS analysis, we determined that the major structural change in oxidized HSA in healthy human plasma is a disulfide-bonded cysteine at the thiol of Cys34 of reduced HSA. Based on this structural information, we prepared standard samples of purified HSA, e.g. nonoxidized (intact purified HSA which mainly exists in reduced form), mildly oxidized and highly oxidized HSA. Using these standards, we demonstrated several differences in functional properties of HSA including protease susceptibility, ligand-binding affinity and antioxidant activity. From these observations, we conclude that an increased level of oxidized HSA may impair HSA function in a number of pathological conditions.


Subject(s)
Antioxidants/metabolism , Serum Albumin/chemistry , Serum Albumin/metabolism , Amino Acid Sequence , Chromatography, Liquid , Computer Simulation , Cysteine/metabolism , Electron Spin Resonance Spectroscopy , Humans , Hydrolysis , Ligands , Models, Molecular , Molecular Sequence Data , Molecular Weight , Oxidation-Reduction , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Serum Albumin/analysis , Serum Albumin/isolation & purification , Spectrometry, Mass, Electrospray Ionization , Spectroscopy, Fourier Transform Infrared , Structure-Activity Relationship , Trypsin/pharmacology
18.
Hepatol Res ; 33(1): 27-32, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16169275

ABSTRACT

BCAA granules (a mixture of branched-chain amino acids) have been used to reverse the hypoalbuminemia of decompensated liver cirrhotic patients in Japan. Our previous studies showed that BCAA promoted albumin secretion through the mTOR signal transduction pathway in rat primary hepatocyte culture [Ijichi C, Matsumura T, Tsuji T, Eto Y. Branched-chain amino acids promote albumin synthesis in rat primary hepatocytes through the mTOR signal transduction system. Biochem Biophys Res Commun 2003;303:59-64]. However, the mTOR-activating effect of BCAA in the experimental cirrhotic animals presenting with hypoalbuminemia has not yet been examined. The purpose of this study is to assess whether oral administration of BCAA induces mTOR activity in the livers of normal rats and CCl(4)-induced cirrhotic rats (CCl(4) rats). Biochemical analysis of liver extracts isolated from several rats showed that oral administration of BCAA (0.75g/kg body weight (BW)) induced phosphorylation of 4E-BP1 and stimulated the enzymatic activity of p70 S6K. Both of these molecules act downstream of mTOR. From the results, we conclude that orally administrated BCAA augments albumin synthesis in the liver, not only by supplementation of material substrates for protein synthesis, but also by induction of an mTOR signal that is critical for translational initiation. Furthermore, we conclude that induction of mTOR signaling is one of the major pharmacological mechanisms by which BCAA granules reverse the hypoalbuminemia of cirrhotic patients.

19.
Am J Physiol Gastrointest Liver Physiol ; 288(6): G1292-300, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15591158

ABSTRACT

It is well established that impaired glucose metabolism is a frequent complication in patients with hepatic cirrhosis. We previously showed that leucine, one of the branched-chain amino acids (BCAA), promotes glucose uptake under insulin-free conditions in isolated skeletal muscle from normal rats. The aim of the present study was to evaluate the effects of BCAA on glucose metabolism in a rat model of CCl(4)-induced cirrhosis (CCl(4) rats). Oral glucose tolerance tests were performed on BCAA-treated CCl(4) rats. In the CCl(4) rats, treatment with leucine or isoleucine, but not valine, improved glucose tolerance significantly, with the effect of isoleucine being greater than the effect of leucine. Glucose uptake experiments using isolated soleus muscle from the CCl(4) rats revealed that leucine and isoleucine, but not valine, promoted glucose uptake under insulin-free conditions. To clarify the mechanism of the blood glucose-lowering effects of BCAA, we collected soleus muscles from BCAA-treated CCl(4) rats with or without a glucose load. These samples were used to determine the subcellular location of glucose transporter proteins and glycogen synthase (GS) activity. Oral administration of leucine or isoleucine without a glucose load induced GLUT4 and GLUT1 translocation to the plasma membrane. GS activity was augmented only in leucine-treated rats and was completely inhibited by rapamycin, an inhibitor of mammalian target of rapamycin. In summary, we found that leucine and isoleucine improved glucose metabolism in CCl(4) rats by promoting glucose uptake in skeletal muscle. This effect occurred as a result of upregulation of GLUT4 and GLUT1 and also by mammalian target of rapamycin-dependent activation of GS in skeletal muscle. From these results, we consider that BCAA treatment may have beneficial effects on glucose metabolism in cirrhotic patients.


Subject(s)
Glucose/metabolism , Isoleucine/pharmacology , Leucine/pharmacology , Liver Cirrhosis/complications , Administration, Oral , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/adverse effects , Disease Models, Animal , Glucose Transporter Type 4 , Glycogen Synthase/pharmacology , Isoleucine/administration & dosage , Leucine/administration & dosage , Liver Cirrhosis/veterinary , Male , Monosaccharide Transport Proteins/pharmacology , Muscle Proteins/pharmacology , Rats , Rats, Sprague-Dawley
20.
Hepatol Res ; 30S: 19-24, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15607134

ABSTRACT

LIVACT granules, which is a branched-chain amino acid (BCAA) preparation, was developed for the purpose of improving hypoalbuminemia in patients with uncompensated liver cirrhosis in Japan. Recent clinical studies have shown that BCAA supplementation not only improves hypoalbuminemia, but also reduces the occurrence frequency of various complications of liver cirrhosis, which considerably affect mortality. In order to comprehend the significance of BCAA supplementation in patients with liver cirrhosis and to suggest better treatments, we conducted basic non-clinical studies mainly using animal models, clarified the molecular mechanism of the curative effect on hypoalbuminemia and emphasized the importance of mTOR signal transduction. Moreover, we found a new pharmacological action of BCAA, which improves glucose metabolism in skeletal muscles.

SELECTION OF CITATIONS
SEARCH DETAIL
...