Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
PLoS One ; 13(11): e0203978, 2018.
Article in English | MEDLINE | ID: mdl-30500824

ABSTRACT

IKKß, an essential kinase of NF-κB signaling, is composed of an N-terminal kinase domain (KD) and a C-terminal scaffolding domain, containing a ubiquitin-like domain (ULD). The Hsp90 chaperon has special responsibility for folding of protein kinases including IKKß. Here, we found that Hsp90 inhibition induced IKKß degradation, which is partially mediated by Keap1. Geldanamycin (GA), a Hsp90 inhibitor, enhances association of IKKß with Keap1 through the binding site in KD, and translocates IKKß to detergent-insoluble fractions leading to its autophagic degradation. An electrophile tBHQ suppressed Keap1-mediated proteasomal Nrf2 degradation but not autophagic IKKß degradation. Substitution mutation of Leu353 to Ala in the ULD destabilizes IKKß, enhances its association with Keap1, translocates it to detergent-insoluble fractions, and causes its autophagic degradation. These results suggest that Keap1 is involved in the degradation of structural destabilized IKKß and negative regulation of NF-κB under proteotoxic stress.


Subject(s)
Autophagy , I-kappa B Kinase/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Proteolysis , Amino Acid Substitution , Animals , HEK293 Cells , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , I-kappa B Kinase/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Mice , Mice, Knockout , Mutation, Missense , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Domains , Protein Folding , Protein Transport/genetics
2.
FEBS Lett ; 591(24): 4083-4094, 2017 12.
Article in English | MEDLINE | ID: mdl-29139553

ABSTRACT

PP2A is composed of a scaffolding subunit (A), a catalytic subunit (C) and a regulatory subunit (B) that is classified into four families including B, B', B'' and B'''/striatin. Here, we found that a distinct PP2A complex regulates NF-κB signalling by dephosphorylation of IKKß, IκBα and RelA/p65. The PP2A core enzyme AC dimer and the holoenzyme AB'''C trimer dephosphorylate IKKß, IκBα and RelA, whereas the ABC trimer dephosphorylates IκBα but not IKKß and RelA in cells. In contrast, AB'C and AB''C trimers have little effect on dephosphorylation of these signalling proteins. These results suggest that different forms of PP2A regulate NF-κB pathway signalling through multiple steps each in a different manner, thereby finely tuning NF-κB- and IKKß-mediated cellular responses.


Subject(s)
I-kappa B Kinase/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Protein Phosphatase 2/physiology , Transcription Factor RelA/metabolism , Cells, Cultured , Humans , Phosphorylation , Protein Subunits/physiology , Signal Transduction
3.
Biochem Biophys Res Commun ; 484(3): 522-528, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28126338

ABSTRACT

Insulin receptor substrates (IRSs) are phosphorylated by IGF-I receptor tyrosine kinase in a ligand-dependent manner. In turn, they bind to and activate effector proteins such as PI3K, leading to various cell responses including cell proliferation. We had reported that ubiquitin ligase Nedd4 induces mono-ubiquitination of IRS-2, thereby enhancing IRS-2 tyrosine phosphorylation, leading to increased IGF signaling and mitogenic activity. Here we show that ubiquitin-specific protease 15 (USP15) antagonizes the effect of Nedd4 on IRS-2. We identified USP15 as a protein that preferentially bound to IRS-2 when IRS-2 was conjugated with ubiquitin. In HEK293 cells, Nedd4 overexpression induced IRS-2 ubiquitination, which was decreased by USP15 co-expression while increased by USP15 knockdown. Nedd4 overexpression enhanced IGF-I-dependent IRS-2 tyrosine phosphorylation, and USP15 co-expression suppressed it. Conversely, USP15 knockdown increased IRS-2 tyrosine phosphorylation and downstream signaling in prostate cancer PC-3 cells. We concluded that USP15 attenuates IGF-I signaling by antagonizing Nedd4-induced IRS-2 ubiquitination.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination/physiology , HEK293 Cells , Humans , Nedd4 Ubiquitin Protein Ligases , Ubiquitinated Proteins/metabolism
4.
Sci Rep ; 6: 20157, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26818807

ABSTRACT

Resistin-like molecule ß (RELMß) reportedly has multiple functions including local immune responses in the gut. In this study, we investigated the possible contribution of RELMß to non-alcoholic steatohepatitis (NASH) development. First, RELMß knock-out (KO) mice were shown to be resistant to methionine-choline deficient (MCD) diet-induced NASH development. Since it was newly revealed that Kupffer cells in the liver express RELMß and that RELMß expression levels in the colon and the numbers of RELMß-positive Kupffer cells were both increased in this model, we carried out further experiments using radiation chimeras between wild-type and RELMß-KO mice to distinguish between the contributions of RELMß in these two organs. These experiments revealed the requirement of RELMß in both organs for full manifestation of NASH, while deletion of each one alone attenuated the development of NASH with reduced serum lipopolysaccharide (LPS) levels. The higher proportion of lactic acid bacteria in the gut microbiota of RELMß-KO than in that of wild-type mice may be one of the mechanisms underlying the lower serum LPS level the former. These data suggest the contribution of increases in RELMß in the gut and Kupffer cells to NASH development, raising the possibility of RELMß being a novel therapeutic target for NASH.


Subject(s)
Choline Deficiency , Diet , Hormones, Ectopic/genetics , Methionine/deficiency , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Biomarkers , Colon/metabolism , Disease Models, Animal , Gastrointestinal Microbiome , Gene Expression Regulation , Hormones, Ectopic/metabolism , Intercellular Signaling Peptides and Proteins , Kupffer Cells/metabolism , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/pathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Transcription, Genetic
5.
J Am Heart Assoc ; 5(1)2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26796253

ABSTRACT

BACKGROUND: The homologous to the E6-AP carboxyl terminus (HECT)-type ubiquitin E3 ligase ITCH is an enzyme that plays a pivotal role in posttranslational modification by ubiquitin proteasomal protein degradation. Thioredoxin-interacting protein (TXNIP) is a negative regulator of the thioredoxin system and an endogenous reactive oxygen species scavenger. In the present study, we focused on the functional role of ubiquitin E3 ligase ITCH and its interaction with TXNIP to elucidate the mechanism of cardiotoxicity induced by reactive oxygen species, such as doxorubicin and hydrogen peroxide. METHODS AND RESULTS: Protein interaction between TXNIP and ITCH in cardiomyocyte was confirmed by immunoprecipitation assays. Overexpression of ITCH increased proteasomal TXNIP degradation and augmented thioredoxin activity, leading to inhibition of reactive oxygen species generation, p38 MAPK, p53, and subsequent intrinsic pathway cardiomyocyte apoptosis in reactive oxygen species-induced cardiotoxicity. Conversely, knockdown of ITCH using small interfering RNA inhibited TXNIP degradation and resulted in a subsequent increase in cardiomyocyte apoptosis. Next, we generated a transgenic mouse with cardiac-specific overexpression of ITCH, called the ITCH-Tg mouse. The expression level of TXNIP in the myocardium in ITCH-Tg mice was significantly lower than WT littermates. In ITCH-Tg mice, cardiac dysfunction and remodeling were restored compared with WT littermates after doxorubicin injection and myocardial infarction surgery. Kaplan-Meier analysis revealed that ITCH-Tg mice had a higher survival rate than WT littermates after doxorubicin injection and myocardial infarction surgery. CONCLUSION: We demonstrated, for the first time, that ITCH targets TXNIP for ubiquitin-proteasome degradation in cardiomyocytes and ameliorates reactive oxygen species-induced cardiotoxicity through the thioredoxin system.


Subject(s)
Cardiomyopathies/enzymology , Carrier Proteins/metabolism , Myocardial Infarction/enzymology , Myocytes, Cardiac/enzymology , Oxidative Stress , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Newborn , Apoptosis , Cardiomyopathies/chemically induced , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cardiotoxicity , Carrier Proteins/genetics , Cell Cycle Proteins , Cells, Cultured , Disease Models, Animal , Doxorubicin , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex , Protein Binding , Proteolysis , RNA Interference , Rats, Sprague-Dawley , Signal Transduction , Thioredoxins/genetics , Time Factors , Transfection , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Ventricular Function, Left , Ventricular Remodeling , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Diabetol Metab Syndr ; 7: 104, 2015.
Article in English | MEDLINE | ID: mdl-26594248

ABSTRACT

BACKGROUND: Insulin resistance with elevated glucose is a risk factor for non-alcoholic steatohepatitis (NASH). We investigated the effects of the sodium glucose cotransporter 2 (SGLT2) inhibitor luseogliflozin on NASH development using a rodent model. METHODS: Mice were treated with both nicotinamide and streptozotocin (NA/STZ) to reduce insulin secretory capacity, and then fed a high fat diet containing trans fatty acids (HFDT) for 8 weeks. The NA/STZ HFDT-fed mice were divided into two groups, either treated with luseogliflozin or untreated, during this period. The glucose elevations in the NA/STZ-treated and HFDT-fed mice were significantly improved by luseogliflozin administration. While HFDT feeding induced NASH development as shown by liver weight gain with lipid accumulation and increased serum alanine aminotransferase, these changes were all attenuated in the group treated with luseogliflozin. In addition, fibrotic change and increases in collagen deposition with upregulations of collagen1 and smooth muscle actin and inflammatory cytokine expressions observed in the HFDT-fed mouse livers were also normalized by luseogliflozin administration. CONCLUSIONS: Taken together, these results obtained in mice demonstrate the favorable effects of administering SGLT2 inhibitors, for the treatment of NASH associated with diabetes mellitus. We anticipate that these agents would be applicable to humans.

7.
J Biol Chem ; 290(40): 24255-66, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26276391

ABSTRACT

AMP-activated protein kinase (AMPK) plays a critical role in metabolic regulation. In this study, first, it was revealed that Pin1 associates with any isoform of γ, but not with either the α or the ß subunit, of AMPK. The association between Pin1 and the AMPK γ1 subunit is mediated by the WW domain of Pin1 and the Thr(211)-Pro-containing motif located in the CBS domain of the γ1 subunit. Importantly, overexpression of Pin1 suppressed AMPK phosphorylation in response to either 2-deoxyglucose or biguanide stimulation, whereas Pin1 knockdown by siRNAs or treatment with Pin1 inhibitors enhanced it. The experiments using recombinant Pin1, AMPK, LKB1, and PP2C proteins revealed that the protective effect of AMP against PP2C-induced AMPKα subunit dephosphorylation was markedly suppressed by the addition of Pin1. In good agreement with the in vitro data, the level of AMPK phosphorylation as well as the expressions of mitochondria-related genes, such as PGC-1α, which are known to be positively regulated by AMPK, were markedly higher with reduced triglyceride accumulation in the muscles of Pin1 KO mice as compared with controls. These findings suggest that Pin1 plays an important role in the pathogenic mechanisms underlying impaired glucose and lipid metabolism, functioning as a negative regulator of AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Phosphatase 2/metabolism , Animals , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Gene Silencing , Glucose/chemistry , HEK293 Cells , Hep G2 Cells , Humans , Lipid Metabolism , Metabolic Syndrome/metabolism , Metformin/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscles/pathology , NIMA-Interacting Peptidylprolyl Isomerase , Phosphorylation , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism
8.
Mediators Inflamm ; 2015: 125380, 2015.
Article in English | MEDLINE | ID: mdl-26170532

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a disorder characterized by hepatic lipid accumulation followed by the inflammation-induced death of hepatocytes and fibrosis. In this process, oxidative stress contributes to the induction of several inflammatory cytokines including TNF-α andIL-1ß in macrophages, while, in hepatocytes, NF-κB reportedly induces the expressions of cell survival genes for protection from apoptosis. Recently, it was reported that the new ubiquitin ligase complex termed linear ubiquitin chain assembly complex (LUBAC), composed of SHARPIN (SHANK-associated RH domain-interacting protein), HOIL-1L (longer isoform of heme-oxidized iron-regulatory protein 2 ubiquitin ligase-1), and HOIP (HOIL-1L interacting protein), forms linear ubiquitin on NF-κB essential modulator (NEMO) and thereby induces NF-κB pathway activation. In this study, we demonstrated the formation of LUBAC to be impaired in the livers of NASH rodent models produced by methionine and choline deficient (MCD) diet feeding, first by either gel filtration or Blue Native-PAGE, with subsequent confirmation by western blotting. The reduction of LUBAC is likely to be attributable to markedly reduced expression of SHARPIN, one of its components. Thus, impaired LUBAC formation, which would result in insufficient NF-κB activation, may be one of the molecular mechanisms underlying the enhanced apoptotic response of hepatocytes in MCD diet-induced NASH livers.


Subject(s)
Choline Deficiency/metabolism , Liver/metabolism , Methionine/deficiency , Non-alcoholic Fatty Liver Disease/metabolism , Ubiquitin/metabolism , Animals , Carrier Proteins/analysis , Carrier Proteins/genetics , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Palmitates/pharmacology , Ubiquitin-Protein Ligases/analysis
9.
PLoS One ; 10(6): e0127467, 2015.
Article in English | MEDLINE | ID: mdl-26039047

ABSTRACT

BACKGROUND: Prostate cancer initially develops in an androgen-dependent manner but, during its progression, transitions to being androgen-independent in the advanced stage. Pin1, one of the peptidyl-prolyl cis/trans isomerases, is reportedly overexpressed in prostate cancers and is considered to contribute to accelerated cell growth, which may be one of the major factors contributing to their androgen-independent growth. Thus, we investigated how Pin1 modulates the gene expressions in both androgen-dependent and androgen-independent prostate cancer cell lines using microarray analysis. In addition, the effects of Juglone, a commercially available Pin1 inhibitor were also examined. METHODS: Two prostate cancer cell-lines, LNCaP (androgen-dependent) and DU145 (androgen-independent), were treated with Pin1 siRNA and its effects on gene expressions were analyzed by microarray. Individual gene regulations induced by Pin1 siRNA or the Pin1 inhibitor Juglone were examined using RT-PCR. In addition, the effects of Juglone on the growth of LNCaP and DU145 transplanted into mice were investigated. RESULTS: Microarray analysis revealed that transcriptional factors regulated by Pin1 differed markedly between LNCaP and DU145 cells, the only exception being that Nrf was regulated in the same way by Pin1 siRNA in both cell lines. Despite this marked difference in gene regulations, Pin1 siRNA and Juglone exert a strong inhibitory effect on both the LNCaP and the DU145 cell line, suppressing in vitro cell proliferation as well as tumor enlargement when transplanted into mice. CONCLUSIONS: Despite Pin1-regulated gene expressions differing between these two prostate cancer cell-lines, LNCaP (androgen-dependent) and DU145 (androgen-independent), Pin1 inhibition suppresses proliferation of both cell-lines. These findings suggest the potential effectiveness of Pin1 inhibitors as therapeutic agents for prostate cancers, regardless of their androgen sensitivity.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Naphthoquinones/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Peptidylprolyl Isomerase/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , NIMA-Interacting Peptidylprolyl Isomerase , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Am J Physiol Endocrinol Metab ; 309(3): E214-23, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26015438

ABSTRACT

Dipeptidyl peptidase IV (DPP-IV) expression in visceral adipose tissue is reportedly increased in obese patients, suggesting an association of DPP-IV with inflammation. In this study, first, lipopolysaccharide (LPS)- or palmitate-induced elevations of inflammatory cytokine mRNA expressions in RAW264.7 macrophages were shown to be significantly suppressed by coincubation with a DPP-IV inhibitor, anagliptin (10 µM), despite low DPP-IV expression in the RAW264.7 cells. Regarding the molecular mechanism, LPS-induced degradation of IκBα and phosphorylations of p65, JNK, and p38, as well as NF-κB and AP-1 promoter activities, were revealed to be suppressed by incubation with anagliptin, indicating suppressive effects of anagliptin on both NF-κB and AP-1 signaling pathways. Anagliptin also acted on 3T3-L1 adipocytes, weakly suppressing the inflammatory cytokine expressions induced by LPS and TNFα. When 3T3-L1 and RAW cells were cocultured and stimulated with LPS, the effects of anagliptin on the suppression of cytokine expressions in 3T3-L1 adipocytes were more marked and became evident at the 10 µM concentration. Anti-inflammatory effects of anagliptin were also observed in vivo on the elevated hepatic and adipose expressions and serum concentrations of inflammatory cytokines in association with the suppression of hepatic NF-κB transcriptional activity in LPS-infused mice. Taking these observations together, the anti-inflammatory properties of anagliptin may be beneficial in terms of preventing exacerbation of diabetes and cardiovascular events.


Subject(s)
Adipocytes, White/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Liver/drug effects , Macrophages/drug effects , NF-kappa B/antagonists & inhibitors , Pyrimidines/pharmacology , 3T3-L1 Cells , Adipocytes, White/immunology , Adipocytes, White/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cell Line, Transformed , Coculture Techniques , Cytokines/agonists , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Liver/immunology , Liver/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Pyrimidines/therapeutic use , Response Elements/drug effects , Signal Transduction/drug effects , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/metabolism , Systemic Inflammatory Response Syndrome/prevention & control
11.
Am J Physiol Gastrointest Liver Physiol ; 309(1): G42-51, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25999428

ABSTRACT

Xanthine oxidase (XO) is an enzyme involved in the production of uric acid (UA) from purine nucleotides. Numerous recent studies have revealed the likelihood of metabolic syndrome including nonalcoholic fatty liver disease (NAFLD) or steatohepatitis (NASH) to be related to hyperuricemia. However, it remains unclear whether elevated serum UA during the development of NAFLD or NASH is a cause or a consequence of these diseases. In this study, the XO inhibitor febuxostat was administered to two types of NASH model mice. Febuxostat exerted a strong protective effect against NASH development induced by a high-fat diet containing trans fatty acid (HFDT). In contrast, methionine choline-deficient-diet-induced NASH development not accompanied by hyperuricemia showed no UA normalization, suggesting that the ameliorating effect of febuxostat occurs via the normalization of hyperuricemia itself and/or accompanying molecular mechanism(s) such as oxidative stress. In the HFDT-fed mice, hyperuricemia, elevated alanine aminotransferase, and increased Tunnel-positive cells in the liver were normalized by febuxostat administration. In addition, upregulation of fatty acid oxidation-related genes, fibrotic change, and increases in collagen deposition, inflammatory cytokine expressions, and lipid peroxidation in the HFDT-fed mice were also normalized by febuxostat administration. Taken together, these observations indicate that administration of febuxostat has a protective effect against HFDT-induced NASH development, suggesting the importance of XO in its pathogenesis. Thus XO inhibitors are potentially potent therapies for patients with NASH, particularly that associated with hyperuricemia.


Subject(s)
Enzyme Inhibitors/pharmacology , Gout Suppressants/pharmacology , Hyperuricemia/drug therapy , Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Thiazoles/pharmacology , Xanthine Oxidase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Choline Deficiency/complications , Cytoprotection , Diet, High-Fat , Febuxostat , Hyperuricemia/blood , Hyperuricemia/enzymology , Hyperuricemia/etiology , Hyperuricemia/pathology , Liver/enzymology , Liver/pathology , Liver Cirrhosis, Experimental/enzymology , Liver Cirrhosis, Experimental/pathology , Liver Cirrhosis, Experimental/prevention & control , Methionine/deficiency , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Uric Acid/blood , Xanthine Oxidase/metabolism
12.
Nat Commun ; 6: 6780, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25879670

ABSTRACT

Insulin-like growth factors (IGFs) induce proliferation of various cell types and play important roles in somatic growth and cancer development. Phosphorylation of insulin receptor substrate (IRS)-1/2 by IGF-I receptor tyrosine kinase is essential for IGF action. Here we identify Nedd4 as an IRS-2 ubiquitin ligase. Nedd4 monoubiquitinates IRS-2, which promotes its association with Epsin1, a ubiquitin-binding protein. Nedd4 recruits IRS-2 to the membrane, probably through promoting Epsin1 binding, and enhances IGF-I receptor-induced IRS-2 tyrosine phosphorylation. In thyroid FRTL-5 cells, activation of the cyclic AMP pathway increases the association of Nedd4 with IRS-2, thereby enhancing IRS-2-mediated signalling and cell proliferation induced by IGF-I. The Nedd4 and IRS-2 association is also required for maximal activation of IGF-I signalling and cell proliferation in prostate cancer PC-3 cells. Nedd4 overexpression accelerates zebrafish embryonic growth through IRS-2 in vivo. We conclude that Nedd4-induced monoubiquitination of IRS-2 enhances IGF signalling and mitogenic activity.


Subject(s)
Cell Proliferation/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Mitosis/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Cell Line, Tumor , Cyclic AMP/metabolism , HEK293 Cells , Humans , Male , Nedd4 Ubiquitin Protein Ligases , Phosphorylation , Prostatic Neoplasms/genetics , Rats , Receptor, IGF Type 1/metabolism , Somatomedins , Zebrafish , Zebrafish Proteins/genetics
13.
Am J Physiol Gastrointest Liver Physiol ; 308(2): G151-8, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25428903

ABSTRACT

Several lines of evidence have suggested a role of gut microbiota in the etiology of nonalcoholic steatohepatitis (NASH). NASH subjects reportedly showed a prolonged orocecal transit time coexistent with small intestinal bacterial overgrowth. We considered the possibility that enhanced gastrointestinal motility would influence gut microbiota and thus investigated the effects of the gastroprokinetic agent mosapride citrate (MC) on gut microbiota and the development of NASH using a methionine-choline deficient (MCD) diet-fed rodent model. Mice were divided into three groups, given the normal chow diet (NCD), the MCD diet, or the MCD diet containing 10 mg·kg(-1)·day(-1) of MC (MCD plus MC) for 6 wk. NASH development was evaluated based on hepatic histochemical findings, serum parameters and various mRNA and/or protein expression levels. MC treatment suppressed MCD diet-induced NASH development, with reduced serum lipopolysaccharide and increased plasma glucagon-like peptide-1 (GLP-1) concentrations. Calculation of the relative abundance of each strain based on gut microbiota analyses indicated lactic acid bacteria specifically, such as Bifidobacterium and Lactobacillus, in feces to be decreased in the MCD, compared with the NCD group. Interestingly, the reduction in lactic acid bacteria in the MCD diet group was reversed in the MCD plus MC group. In addition, colon inflammation observed in the MCD diet group was reduced in the MCD plus MC group. Therefore, MC showed a protective effect against MCD diet-induced NASH development in our rodent model, with possible involvements of increased fecal lactic acid bacteria, protection against colon inflammation and elevated plasma GLP-1.


Subject(s)
Benzamides/pharmacology , Feces/microbiology , Glucagon-Like Peptide 1/blood , Inflammation/metabolism , Lactic Acid/metabolism , Liver/drug effects , Morpholines/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Choline Deficiency/metabolism , Feces/chemistry , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Liver/metabolism , Liver Cirrhosis/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism
14.
Am J Physiol Gastrointest Liver Physiol ; 305(12): G911-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24113768

ABSTRACT

Gut microbiota alterations are associated with various disorders. In this study, gut microbiota changes were investigated in a methionine-choline-deficient (MCD) diet-induced nonalcoholic steatohepatitis (NASH) rodent model, and the effects of administering Lactobacillus casei strain Shirota (LcS) on the development of NASH were also investigated. Mice were divided into three groups, given the normal chow diet (NCD), MCD diet, or the MCD diet plus daily oral administration of LcS for 6 wk. Gut microbiota analyses for the three groups revealed that lactic acid bacteria such as Bifidobacterium and Lactobacillus in feces were markedly reduced by the MCD diet. Interestingly, oral administration of LcS to MCD diet-fed mice increased not only the L. casei subgroup but also other lactic acid bacteria. Subsequently, NASH development was evaluated based on hepatic histochemical findings, serum parameters, and various mRNA and/or protein expression levels. LcS intervention markedly suppressed MCD-diet-induced NASH development, with reduced serum lipopolysaccharide concentrations, suppression of inflammation and fibrosis in the liver, and reduced colon inflammation. Therefore, reduced populations of lactic acid bacteria in the colon may be involved in the pathogenesis of MCD diet-induced NASH, suggesting normalization of gut microbiota to be effective for treating NASH.


Subject(s)
Fatty Liver , Gastrointestinal Tract , Lacticaseibacillus casei/metabolism , Microbiota/physiology , Animals , Bifidobacterium/isolation & purification , Bifidobacterium/metabolism , Choline Deficiency/metabolism , Disease Models, Animal , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/prevention & control , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Gene Expression Profiling/methods , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism , Liver/metabolism , Liver/pathology , Methionine/deficiency , Methionine/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease
15.
Adipocyte ; 2(1): 28-32, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23700549

ABSTRACT

Inflammation involving adipose tissue is regarded as one of the major molecular mechanisms underlying obesity-related insulin resistance. Recent studies have suggested a series of angiotensin II receptor blockers (ARBs) to improve insulin resistance or protect against the development of diabetes mellitus. We previously demonstrated that valsartan suppresses the inflammatory response of macrophages. Interestingly, however, this effect did not occur via peroxisome proliferator-activated receptor (PPAR) γ or the AT1a receptor. This suppression appears to secondarily lead to amelioration of insulin resistance and reductions in abnormal gene expressions in adipocytes. In addition to these in vitro findings, we herein demonstrate the in vivo effects of valsartan, using mice constitutively infused with lipopolysaccharide (LPS) for 4 weeks. Oral administration of valsartan to LPS-infused mice normalized the increased expressions of inflammatory cytokines in adipose and liver tissues. These results raise the possibility that valsartan not only contributes to normalization of obesity-related insulin resistance, but is also beneficial for the treatment of other diseases with inflammation related to the metabolic syndrome such as atherosclerosis and non-alcoholic steatohepatitis. Further study is necessary to clarify these issues.

16.
Arterioscler Thromb Vasc Biol ; 33(8): 1986-93, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23702657

ABSTRACT

OBJECTIVE: Resistin-like molecule (RELM) ß is a secretory protein homologous to resistin and reportedly contributes to local immune response regulation in gut and bronchial epithelial cells. However, we found that activated macrophages also express RELMß and thus investigated the role of RELMß in the development of atherosclerosis. APPROACH AND RESULTS: It was demonstrated that foam cells in atherosclerotic lesions of the human coronary artery abundantly express RELMß. RELMß knockout ((-/-)) and wild-type mice were mated with apolipoprotein E-deficient background mice. RELMß(-/-) apolipoprotein E-deficient mice exhibited less lipid accumulation in the aortic root and wall than RELMß(+/+) apolipoprotein E-deficient mice, without significant changes in serum lipid parameters. In vitro, RELMß(-/-) primary cultured peritoneal macrophages (PCPMs) exhibited weaker lipopolysaccharide-induced nuclear factor-κB classical pathway activation and inflammatory cytokine secretion than RELMß(+/+), whereas stimulation with RELMß upregulated inflammatory cytokine expressions and increased expressions of many lipid transporters and scavenger receptors in PCPMs. Flow cytometric analysis revealed inflammatory stimulation-induced RELMß in F4/80(+) CD11c(+) PCPMs. In contrast, the expressions of CD11c and tumor necrosis factor were lower in RELMß(-/-) PCPMs, but both were restored by stimulation with recombinant RELMß. CONCLUSIONS: RELMß is abundantly expressed in foam cells within plaques and contributes to atherosclerosis development via lipid accumulation and inflammatory facilitation.


Subject(s)
Atherosclerosis/metabolism , Foam Cells/metabolism , Hormones, Ectopic/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , CD11c Antigen/metabolism , Cell Line , Fatty Acids/pharmacology , Female , Foam Cells/immunology , Foam Cells/pathology , Hormones, Ectopic/genetics , Hormones, Ectopic/immunology , Humans , Intercellular Signaling Peptides and Proteins/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Knockout , Primary Cell Culture , Vasculitis/immunology , Vasculitis/metabolism , Vasculitis/pathology
17.
J Biol Chem ; 288(28): 20692-701, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23720771

ABSTRACT

Pin1 and Par14 are parvulin-type peptidyl-prolyl cis/trans isomerases. Although numerous proteins have been identified as Pin1 substrates, the target proteins of Par14 remain largely unknown. Par14 expression levels are increased in the livers and embryonic fibroblasts of Pin1 KO mice, suggesting a compensatory relationship between the functions of Pin1 and Par14. In this study, the association of Par14 with insulin receptor substrate 1 (IRS-1) was demonstrated in HepG2 cells overexpressing both as well as endogenously in the mouse liver. The analysis using deletion-mutated Par14 and IRS-1 constructs revealed the N-terminal portion containing the basic domain of Par14 and the two relatively C-terminal portions of IRS-1 to be involved in these associations, in contrast to the WW domain of Pin1 and the SAIN domain of IRS-1. Par14 overexpression in HepG2 markedly enhanced insulin-induced IRS-1 phosphorylation and its downstream events, PI3K binding with IRS-1 and Akt phosphorylation. In contrast, treating HepG2 cells with Par14 siRNA suppressed these events. In addition, overexpression of Par14 in the insulin-resistant ob/ob mouse liver by adenoviral transfer significantly improved hyperglycemia with normalization of hepatic PEPCK and G6Pase mRNA levels, and gene suppression of Par14 using shRNA adenovirus significantly exacerbated the glucose intolerance in Pin1 KO mice. Therefore, although Pin1 and Par14 associate with different portions of IRS-1, the prolyl cis/trans isomerization in multiple sites of IRS-1 by these isomerases appears to be critical for efficient insulin receptor-induced IRS-1 phosphorylation. This process is likely to be one of the major mechanisms regulating insulin sensitivity and also constitutes a potential therapeutic target for novel insulin-sensitizing agents.


Subject(s)
Insulin Receptor Substrate Proteins/metabolism , Insulin/pharmacology , Peptidylprolyl Isomerase/metabolism , Animals , Binding Sites/genetics , Glucose Intolerance/genetics , HEK293 Cells , Hep G2 Cells , Humans , Hyperglycemia/genetics , Hyperglycemia/therapy , Hypoglycemic Agents/pharmacology , Immunoblotting , Insulin Receptor Substrate Proteins/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mutation , NIMA-Interacting Peptidylprolyl Isomerase , Obesity/blood , Obesity/genetics , Peptidylprolyl Isomerase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference
18.
Biochem Biophys Res Commun ; 434(2): 197-202, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23523797

ABSTRACT

The dynamic process of adipose differentiation involves stepwise expressions of transcription factors and proteins specific to the mature fat cell phenotype. In this study, it was revealed that expression levels of IntS6 and IntS11, subunits of the Integrator complex, were increased in 3T3-L1 cells in the period when the cells reached confluence and differentiated into adipocytes, while being reduced to basal levels after the completion of differentiation. Suppression of IntS6 or IntS11 expression using siRNAs in 3T3-L1 preadipocytes markedly inhibited differentiation into mature adipocytes, based on morphological findings as well as mRNA analysis of adipocyte-specific genes such as Glut4, perilipin and Fabp4. Although Pparγ2 protein expression was suppressed in IntS6 or IntS11-siRNA treated cells, adenoviral forced expression of Pparγ2 failed to restore the capacity for differentiation into mature adipocytes. Taken together, these findings demonstrate that increased expression of Integrator complex subunits is an indispensable event in adipose differentiation. Although further study is necessary to elucidate the underlying mechanism, the processing of U1, U2 small nuclear RNAs may be involved in cell differentiation steps.


Subject(s)
Adipocytes/cytology , Adipogenesis , DEAD-box RNA Helicases/metabolism , Multiprotein Complexes/metabolism , 3T3-L1 Cells , Adenoviridae/metabolism , Adipocytes/metabolism , Animals , Blotting, Western , DEAD-box RNA Helicases/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Mice , Multiprotein Complexes/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins , Time Factors , Transfection , Tumor Suppressor Proteins
19.
J Biol Chem ; 287(53): 44526-35, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23112047

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a disorder characterized by simultaneous fat accumulation and chronic inflammation in the liver. In this study, Pin1 expression was revealed to be markedly increased in the livers of mice with methionine choline-deficient (MCD) diet-induced NASH, a rodent model of NASH. In addition, Pin1 KO mice were highly resistant to MCD-induced NASH, based on a series of data showing simultaneous fat accumulation, chronic inflammation, and fibrosis in the liver. In terms of Pin1-induced fat accumulation, it was revealed that the expression levels of peroxisome proliferator-activated receptor α and its target genes were higher in the livers of Pin1 KO mice than in controls. Thus, resistance of Pin1 KO mice to hepatic steatosis is partially attributable to the lack of Pin1-induced down-regulation of peroxisome proliferator-activated receptor α, although multiple other mechanisms are apparently involved. Another mechanism involves the enhancing effect of hematopoietic Pin1 on the expressions of inflammatory cytokines such as tumor necrosis factor and monocyte chemoattractant protein 1 through NF-κB activation, eventually leading to hepatic fibrosis. Finally, to distinguish the roles of hematopoietic or nonhematopoietic Pin1 in NASH development, mice lacking Pin1 in either nonhematopoietic or hematopoietic cells were produced by bone marrow transplantation between wild-type and Pin1 KO mice. The mice having nonhematopoietic Pin1 exhibited fat accumulation without liver fibrosis on the MCD diet. Thus, hepatic Pin1 appears to be directly involved in the fat accumulation in hepatocytes, whereas Pin1 in hematopoietic cells contributes to inflammation and fibrosis. In summary, this is the first study to demonstrate that Pin1 plays critical roles in NASH development. This report also raises the possibility that hepatic Pin1 inhibition to the appropriate level might provide a novel therapeutic strategy for NASH.


Subject(s)
Fatty Liver/enzymology , Peptidylprolyl Isomerase/metabolism , Animals , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/pathology , Female , Humans , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Non-alcoholic Fatty Liver Disease , Peptidylprolyl Isomerase/genetics
20.
Arterioscler Thromb Vasc Biol ; 32(2): 291-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22095983

ABSTRACT

OBJECTIVE: Hyperuricemia is common in patients with metabolic syndrome. We investigated the role of xanthine oxidoreductase (XOR) in atherosclerosis development, and the effects of the XOR inhibitor allopurinol on this process. METHODS AND RESULTS: Oral administration of allopurinol to ApoE knockout mice markedly ameliorated lipid accumulation and calcification in the aorta and aortic root. In addition, allopurinol treatment or siRNA-mediated gene knockdown of XOR suppressed transformation of J774.1 murine macrophage cells, treated with acetylated LDL or very low density lipoprotein (VLDL) into foam cells. This inhibitory effect of allopurinol was also observed in primary cultured human macrophages. In contrast, overexpression of XOR promoted transformation of J774.1 cells into foam cells. Interestingly, SR-A1, SR-B1, SR-B II, and VLDL receptors in J774.1 cells were reduced by XOR knockdown, and increased by XOR overexpression. Conversely, expressions of ABCA1 and ABCG1 were increased by XOR knockdown and suppressed by XOR overexpression. Finally, productions of inflammatory cytokines accompanied by foam cell formation were also reduced by allopurinol administration. CONCLUSIONS: These results strongly suggest XOR activity and/or its expression level to contribute to macrophage foam cell formation. Thus, XOR inhibitors may be useful for preventing atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Cell Differentiation/physiology , Foam Cells/pathology , Macrophages/pathology , Xanthine Dehydrogenase/physiology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/metabolism , Allopurinol/pharmacology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Foam Cells/drug effects , Foam Cells/metabolism , Humans , Lipid Metabolism/drug effects , Lipoproteins/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Xanthine Dehydrogenase/antagonists & inhibitors , Xanthine Dehydrogenase/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...