Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
J Biol Chem ; 299(3): 102964, 2023 03.
Article in English | MEDLINE | ID: mdl-36736425

ABSTRACT

Cardiac hypertrophy is a crucial risk factor for hypertensive disorders during pregnancy, but its progression during pregnancy remains unclear. We previously showed cardiac hypertrophy in a pregnancy-associated hypertensive (PAH) mouse model, in which an increase in angiotensin II (Ang II) levels was induced by human renin and human angiotensinogen, depending on pregnancy conditions. Here, to elucidate the factors involved in the progression of cardiac hypertrophy, we performed a comprehensive analysis of changes in gene expression in the hearts of PAH mice and compared them with those in control mice. We found that alpha-1A adrenergic receptor (Adra1a) mRNA levels in the heart were significantly reduced under PAH conditions, whereas the renin-angiotensin system was upregulated. Furthermore, we found that Adra1a-deficient PAH mice exhibited more severe cardiac hypertrophy than PAH mice. Our study suggests that Adra1a levels are regulated by renin-angiotensin system and that changes in Adra1a expression are involved in progressive cardiac hypertrophy in PAH mice.


Subject(s)
Angiotensin II , Hypertension, Pregnancy-Induced , Receptors, Adrenergic, alpha-1 , Animals , Female , Humans , Mice , Pregnancy , Angiotensin II/metabolism , Cardiomegaly/metabolism , Myocardium/metabolism , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Renin-Angiotensin System , Hypertension, Pregnancy-Induced/genetics , Hypertension, Pregnancy-Induced/metabolism
2.
Int J Mol Sci ; 25(3)2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38339104

ABSTRACT

One of the extracellular matrix proteins, tenascin-C (TN-C), is known to be upregulated in age-related inflammatory diseases such as cancer and cardiovascular diseases. Expression of this molecule is frequently detected, especially in the macrophage-rich areas of atherosclerotic lesions; however, the role of TN-C in mechanisms underlying the progression of atherosclerosis remains obscure. Previously, we found a hidden bioactive sequence termed TNIIIA2 in the TN-C molecule and reported that the exposure of this sequence would be carried out through limited digestion of TN-C by inflammatory proteases. Thus, we hypothesized that some pro-atherosclerotic phenotypes might be elicited from macrophages when they were stimulated by TNIIIA2. In this study, TNIIIA2 showed the ability to accelerate intracellular lipid accumulation in macrophages. In this experimental condition, an elevation of phagocytic activity was observed, accompanied by a decrease in the expression of transporters responsible for lipid efflux. All these observations were mediated through the induction of excessive ß1-integrin activation, which is a characteristic property of the TNIIIA2 sequence. Finally, we demonstrated that the injection of a drug that targets TNIIIA2's bioactivity could rescue mice from atherosclerotic plaque expansion. From these observations, it was shown that TN-C works as a pro-atherosclerotic molecule through an internal TNIIIA2 sequence. The possible advantages of clinical strategies targeting TNIIIA2 are also indicated.


Subject(s)
Atherosclerosis , Foam Cells , Plaque, Atherosclerotic , Animals , Mice , Extracellular Matrix Proteins , Fibronectins/metabolism , Foam Cells/metabolism , Lipids , Peptides/chemistry , Tenascin/metabolism
3.
Am J Physiol Gastrointest Liver Physiol ; 323(6): G627-G639, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36283088

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) are master transcription factors for lipid synthesis, and SREBP-1 is important for fatty acid and triglyceride synthesis. SREBP-1 has two isoforms, SREBP-1a and SREBP-1c, which are splicing variants transcribed from the Srebf1 gene. Although SREBP-1a exhibits stronger transcriptional activity than SREBP-1c, hepatic SREBP-1c is considered more physiologically important. We generated SREBP-1a flox mice using the CRISPR/Cas9 system and hepatocyte- and macrophage-specific SREBP-1a knockout (KO) mice (LKO, liver-knockout; and mΦKO, macrophage-knockout). There were no significant differences among all the mouse genotypes upon feeding with a normal diet. However, feeding with a methionine- and choline-deficient (MCD) diet resulted in exacerbated liver injury in both KO mice. In LKO mice, fatty liver was unexpectedly exacerbated, leading to macrophage infiltration and inflammation. In contrast, in mΦKO mice, the fatty liver state was similar to that in flox mice, but the polarity of the macrophages in the liver was transformed into a proinflammatory M1 subtype, resulting in the exacerbation of inflammation. Taken together, we found that SREBP-1a does not contribute to hepatic lipogenesis, but in either hepatocytes or macrophages distinctly controls the onset of pathological conditions in MCD diet-induced hepatitis.NEW & NOTEWORTHY Hepatocyte- and macrophage-specific SREBP-1a knockout mice were generated for the first time. This study reveals that SREBP-1a does not contribute to hepatic lipogenesis, but in either hepatocytes or macrophages distinctly controls the onset of pathological conditions in methionine- and choline-deficient diet-induced hepatitis.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Methionine , Choline/metabolism , Mice, Inbred C57BL , Hepatocytes/metabolism , Liver/metabolism , Mice, Knockout , Diet/adverse effects , Inflammation/metabolism , Macrophages/metabolism
4.
FASEB J ; 35(6): e21663, 2021 06.
Article in English | MEDLINE | ID: mdl-34042217

ABSTRACT

cAMP responsive element-binding protein H (CREBH) is a hepatic transcription factor to be activated during fasting. We generated CREBH knock-in flox mice, and then generated liver-specific CREBH transgenic (CREBH L-Tg) mice in an active form. CREBH L-Tg mice showed a delay in growth in the postnatal stage. Plasma growth hormone (GH) levels were significantly increased in CREBH L-Tg mice, but plasma insulin-like growth factor 1 (IGF1) levels were significantly decreased, indicating GH resistance. In addition, CREBH overexpression significantly increased hepatic mRNA and plasma levels of FGF21, which is thought to be as one of the causes of growth delay. However, the additional ablation of FGF21 in CREBH L-Tg mice could not correct GH resistance at all. CREBH L-Tg mice sustained GH receptor (GHR) reduction and the increase of IGF binding protein 1 (IGFBP1) in the liver regardless of FGF21. As GHR is a first step in GH signaling, the reduction of GHR leads to impairment of GH signaling. These data suggest that CREBH negatively regulates growth in the postnatal growth stage via various pathways as an abundant energy response by antagonizing GH signaling.


Subject(s)
Body Composition , Body Mass Index , Cyclic AMP Response Element-Binding Protein/physiology , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental , Growth Hormone/metabolism , Liver/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction
5.
Biochem Biophys Res Commun ; 562: 146-153, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34052660

ABSTRACT

While molecular oxygen is essential for aerobic organisms, its utilization is inseparably connected with generation of oxidative insults. To cope with the detrimental aspects, cells evolved antioxidative defense systems, and insufficient management of the oxidative insults underlies the pathogenesis of a wide range of diseases. A battery of genes for this antioxidative defense are regulated by the transcription factors nuclear factor-erythroid 2-like 1 and 2 (NRF1 and NRF2). While the regulatory steps for the activation of NRFs have been investigated with particular emphasis on nuclear translocation and proteosomal degradation, unknown redundancy may exist considering the indispensable nature of these defense systems. Here we unraveled that C-terminal binding protein 2 (CtBP2), a transcriptional cofactor with redox-sensing capability, is an obligate partner of NRFs. CtBP2 forms transcriptional complexes with NRF1 and NRF2 that is required to promote the expression of antioxidant genes in response to oxidative insults. Our findings illustrate a basis for understanding the transcriptional regulation of antioxidative defense systems that may be exploited therapeutically.


Subject(s)
Alcohol Oxidoreductases/metabolism , Co-Repressor Proteins/metabolism , NF-E2-Related Factor 1/metabolism , NF-E2-Related Factor 2/metabolism , Amino Acid Sequence , Antioxidants/metabolism , Gene Expression Regulation , Humans , NF-E2-Related Factor 1/chemistry , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 2/chemistry , NF-E2-Related Factor 2/genetics , Oxidative Stress , Protein Binding , Transcription, Genetic
6.
Hepatology ; 71(5): 1609-1625, 2020 05.
Article in English | MEDLINE | ID: mdl-31529722

ABSTRACT

BACKGROUND AND AIMS: Dysfunctional hepatic lipid metabolism is a cause of nonalcoholic fatty liver disease (NAFLD), the most common chronic liver disorder worldwide, and is closely associated with insulin resistance and type 2 diabetes. ELOVL fatty acid elongase 6 (Elovl6) is responsible for converting C16 saturated and monounsaturated fatty acids (FAs) into C18 species. We have previously shown that Elovl6 contributes to obesity-induced insulin resistance by modifying hepatic C16/C18-related FA composition. APPROACH AND RESULTS: To define the precise molecular mechanism by which hepatic Elovl6 affects energy homeostasis and metabolic disease, we generated liver-specific Elovl6 knockout (LKO) mice. Unexpectedly, LKO mice were not protected from high-fat diet-induced insulin resistance. Instead, LKO mice exhibited higher insulin sensitivity than controls when consuming a high-sucrose diet (HSD), which induces lipogenesis. Hepatic patatin-like phospholipase domain-containing protein 3 (Pnpla3) expression was down-regulated in LKO mice, and adenoviral Pnpla3 restoration reversed the enhancement in insulin sensitivity in HSD-fed LKO mice. Lipidomic analyses showed that the hepatic ceramide(d18:1/18:0) content was lower in LKO mice, which may explain the effect on insulin sensitivity. Ceramide(d18:1/18:0) enhances protein phosphatase 2A (PP2A) activity by interfering with the binding of PP2A to inhibitor 2 of PP2A, leading to Akt dephosphorylation. Its production involves the formation of an Elovl6-ceramide synthase 4 (CerS4) complex in the endoplasmic reticulum and a Pnpla3-CerS4 complex on lipid droplets. Consistent with this, liver-specific Elovl6 deletion in ob/ob mice reduced both hepatic ceramide(d18:1/18:0) and PP2A activity and ameliorated insulin resistance. CONCLUSIONS: Our study demonstrates the key role of hepatic Elovl6 in the regulation of the acyl-chain composition of ceramide and that C18:0-ceramide is a potent regulator of hepatic insulin signaling linked to Pnpla3-mediated NAFLD.


Subject(s)
Ceramides/metabolism , Fatty Acid Elongases/physiology , Insulin Resistance/genetics , Liver/enzymology , Animals , Ceramides/chemistry , Dietary Sucrose/administration & dosage , Down-Regulation , Fatty Acid Elongases/genetics , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Phospholipases A2, Calcium-Independent/metabolism , Protein Phosphatase 2/metabolism , Sphingosine N-Acyltransferase/metabolism
7.
Biochem Biophys Res Commun ; 523(2): 354-360, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31866012

ABSTRACT

The epithelial to mesenchymal transition (EMT) is a cell intrinsic program controlling cellular morphological and phenotypic remodeling in a wide range of biological processes. Despite the accumulating evidence, the transcriptional networks regulating EMT still remain to be elucidated. In this study, we demonstrate that C-terminal binding protein 2 (CtBP2), a critical transcriptional co-repressor harboring pyridine nucleotide sensing capability, orchestrates the EMT program at least in part through a novel transcriptional interaction with an octamer transcription factor, OCT1 (POU2F1, POU class 2 homeobox 1). We identified novel interactions of CtBP2 with several octamer transcription factors, and CtBP2 exhibits a direct interaction with OCT1 in particular. OCT1 accelerates the EMT program as reported, which is diminished by the mutation of the CtBP-binding motif in OCT1, suggesting OCT1 represses epithelial gene expression through recruiting the co-repressor CtBP2. In accordance with these findings, a canonical EMT activator transforming growth factor-ß (TGF-ß) promotes the formation of the CtBP2/OCT1 complex. Our observations illustrate the role of CtBP2 to orchestrate the EMT program through the interaction with OCT1 and highlight the potential of therapeutic exploitation of this new transcriptional system for a wide range of diseases.


Subject(s)
Alcohol Oxidoreductases/metabolism , Co-Repressor Proteins/metabolism , Epithelial-Mesenchymal Transition/physiology , Octamer Transcription Factor-1/metabolism , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/genetics , Conserved Sequence , Epithelial-Mesenchymal Transition/genetics , Female , Gene Regulatory Networks , Humans , MCF-7 Cells , Mice , Mutation , Octamer Transcription Factor-1/chemistry , Octamer Transcription Factor-1/genetics , Protein Interaction Domains and Motifs , Rats , Transforming Growth Factor beta/metabolism
8.
Int J Mol Sci ; 21(1)2020 Jan 06.
Article in English | MEDLINE | ID: mdl-31935812

ABSTRACT

The selective PPARα modulator (SPPARMα) is expected to medicate dyslipidemia with minimizing adverse effects. Recently, pemafibrate was screened from the ligand library as an SPPARMα bearing strong potency. Several clinical pieces of evidence have proved the usefulness of pemafibrate as a medication; however, how pemafibrate works as a SPPARMα at the molecular level is not fully known. In this study, we investigate the molecular mechanism behind its novel SPPARMα character through a combination of approaches of X-ray crystallography, isothermal titration calorimetry (ITC), and fragment molecular orbital (FMO) analysis. ITC measurements have indicated that pemafibrate binds more strongly to PPARα than to PPARγ. The crystal structure of PPARα-ligand binding domain (LBD)/pemafibrate/steroid receptor coactivator-1 peptide (SRC1) determined at 3.2 Å resolution indicates that pemafibrate binds to the ligand binding pocket (LBP) of PPARα in a Y-shaped form. The structure also reveals that the conformation of the phenoxyalkyl group in pemafibrate is flexible in the absence of SRC1 coactivator peptide bound to PPARα; this gives a freedom for the phenoxyalkyl group to adopt structural changes induced by the binding of coactivators. FMO calculations have indicated that the accumulation of hydrophobic interactions provided by the residues at the LBP improve the interaction between pemafibrate and PPARα compared with the interaction between fenofibrate and PPARα.


Subject(s)
Benzoxazoles/pharmacology , Butyrates/pharmacology , Molecular Docking Simulation , PPAR alpha/chemistry , Benzoxazoles/chemistry , Binding Sites , Butyrates/chemistry , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , PPAR alpha/metabolism , Protein Binding
9.
Int J Mol Sci ; 20(15)2019 Jul 27.
Article in English | MEDLINE | ID: mdl-31357643

ABSTRACT

Obesity causes various health problems, such as type 2 diabetes, non-alcoholic fatty liver disease, and cardio- and cerebrovascular diseases. Metabolic organs, particularly white adipose tissue (WAT) and liver, are deeply involved in obesity. WAT contains many adipocytes with energy storage capacity and secretes adipokines depending on the obesity state, while liver plays pivotal roles in glucose and lipid metabolism. This review outlines and underscores the relationship between obesity and lysosomal functions, including lysosome biogenesis, maturation and activity of lysosomal proteases in WAT and liver. It has been revealed that obesity-induced abnormalities of lysosomal proteases contribute to inflammation and cellular senescence in adipocytes. Previous reports have demonstrated obesity-induced ectopic lipid accumulation in liver is associated with abnormality of lysosomal proteases as well as other lysosomal enzymes. These studies demonstrate that lysosomal dysfunction in WAT and liver underlies part of the obesity-related pathology, raising the possibility that strategies to modulate lysosomal function may be effective in preventing or treating the metabolic syndrome.


Subject(s)
Disease Susceptibility , Lysosomes/metabolism , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Obesity/etiology , Obesity/metabolism , Adipose Tissue/metabolism , Animals , Humans , Liver/metabolism , Metabolic Syndrome/prevention & control , Peptide Hydrolases/metabolism
10.
Biochem Biophys Res Commun ; 499(2): 239-245, 2018 05 05.
Article in English | MEDLINE | ID: mdl-29567478

ABSTRACT

Peroxisome proliferator-activated receptor-α (PPARα) is a ligand-activated transcription factor involved in the regulation of lipid homeostasis and improves hypertriglyceridemia. Pemafibrate is a novel selective PPARα modulator (SPPARMα) that activates PPARα transcriptional activity. Here, we computationally constructed the structure of the human PPARα in a complex with pemafibrate, along with that of hPPARα complexed with the classical fenofibrate, and studied their interactions quantitatively by using the first-principles calculations-based fragment molecular orbital (FMO) method. Comprehensive structural and protein-ligand binding elucidation along with the in vitro luciferase analysis let us to identify pemafibrate as a novel SPPARMα. Unlike known fibrate ligands, which bind only with the arm I of the Y-shaped ligand binding pocket, the Y-shaped pemafibrate binds to the entire cavity region. This lock and key nature causes enhanced induced fit in pemafibrate-ligated PPARα. Importantly, this selective modulator allosterically changes PPARα conformation to form a brand-new interface, which in turn binds to PPARα co-activator, PGC-1α, resulting in the full activation of PPARα. The structural basis for the potent effects of pemafibrate on PPARα transcriptional activity predicted by the in silico FMO methods was confirmed by in vitro luciferase assay for mutants. The unique binding mode of pemafibrate reveals a new pattern of nuclear receptor ligand recognition and suggests a novel basis for ligand design, offering cues for improving the binding affinity and selectivity of ligand for better clinical consequences. The findings explain the high affinity and efficacy of pemafibrate, which is expected to be in the clinical use soon.


Subject(s)
Benzoxazoles/chemistry , Benzoxazoles/metabolism , Butyrates/chemistry , Butyrates/metabolism , Models, Molecular , PPAR alpha/chemistry , PPAR alpha/metabolism , Fenofibrate/chemistry , Fenofibrate/metabolism , Hep G2 Cells , Humans , Ligands , Luciferases/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
11.
Hepatology ; 66(5): 1430-1443, 2017 11.
Article in English | MEDLINE | ID: mdl-28621467

ABSTRACT

Mechanisms of hepatic fibrogenesis induced by hepatitis C virus (HCV), one of the leading causes of liver fibrosis, are not fully understood. We studied transcriptional up-regulation of transforming growth factor ß (TGF-ß), especially TGF-ß2, which is mediated by activation of liver-enriched transcription factor cAMP-responsive element-binding protein, hepatocyte specific (CREBH) triggered by HCV infection and its functional significance for induction of profibrogenic phenotypes by interaction of HCV-infected cells with hepatic stellate cells (HSCs). Compared to TGF-ß1, expression of TGF-ß2 mRNA was induced faster and to a higher level upon HCV infection. Serum TGF-ß2 levels in hepatitis C patients were higher compared to those in healthy individuals and were positively correlated with hepatic fibrosis stages F0-F2. TGF-ß2 promoter activity was decreased and increased, respectively, by silencing and overexpression of CREBH. CREBH recognition sites were identified in the TGF-ß2 promoter. CREBH binding to the promoter and its increase in cells expressing HCV Core-NS2 were shown by gel mobility shift and chromatin immunoprecipitation, respectively. The active form of CREBH was detectable in HCV-infected chimeric mice with human livers and cells expressing HCV proteins. Involvement of CREBH in HCV-induced fibrogenic response was further demonstrated in the CREBH null-mutant mouse model. Fibrogenic phenotypes were assessed using co-cultures of HCV-infected cells and HSCs. Expressions of fibrogenic factors and TGF-ß1 increasing in the co-cultures was prevented by TGF-ß2- or CREBH silencing. CONCLUSION: CREBH was identified as a key positive regulator of TGF-ß2 transcription in HCV-infected cells. TGF-ß2 released from infected cells potentially contributes to cross-induction of TGF-ß in an autocrine manner through its own signaling pathway, leading to an increase in fibrogenic responses in adjacent HSCs. (Hepatology 2017;66:1430-1443).


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Hepatitis C/metabolism , Liver Cirrhosis/virology , Liver/pathology , Transforming Growth Factor beta2/metabolism , Animals , Autocrine Communication , Fibrosis , Gene Expression Regulation , Hepatic Stellate Cells/pathology , Hepatitis C/complications , Hepatitis C/pathology , Liver Cirrhosis/metabolism , Male , Mice, Inbred C57BL , Paracrine Communication , Transforming Growth Factor beta1/metabolism
12.
Int J Mol Sci ; 19(5)2018 May 08.
Article in English | MEDLINE | ID: mdl-29738435

ABSTRACT

The cyclic adenosine monophosphate (cAMP)-responsive element-binding protein H (CREBH, encoded by CREB3L3) is a membrane-bound transcriptional factor that primarily localizes in the liver and small intestine. CREBH governs triglyceride metabolism in the liver, which mediates the changes in gene expression governing fatty acid oxidation, ketogenesis, and apolipoproteins related to lipoprotein lipase (LPL) activation. CREBH in the small intestine reduces cholesterol transporter gene Npc1l1 and suppresses cholesterol absorption from diet. A deficiency of CREBH in mice leads to severe hypertriglyceridemia, fatty liver, and atherosclerosis. CREBH, in synergy with peroxisome proliferator-activated receptor α (PPARα), has a crucial role in upregulating Fgf21 expression, which is implicated in metabolic homeostasis including glucose and lipid metabolism. CREBH binds to and functions as a co-activator for both PPARα and liver X receptor alpha (LXRα) in regulating gene expression of lipid metabolism. Therefore, CREBH has a crucial role in glucose and lipid metabolism in the liver and small intestine.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Glucose/metabolism , Lipid Metabolism/genetics , Animals , Apolipoproteins/genetics , Apolipoproteins/metabolism , Humans , Intestine, Small/metabolism , Lipoprotein Lipase/genetics , Liver/metabolism , Liver X Receptors/genetics , Mice
13.
Int J Mol Sci ; 19(7)2018 Jul 23.
Article in English | MEDLINE | ID: mdl-30041488

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) is a therapeutic target for hyperlipidemia. Pemafibrate (K-877) is a new selective PPARα modulator activating PPARα transcriptional activity. To determine the effects of pemafibrate on diet-induced obesity, wild-type mice were fed a high-fat diet (HFD) containing pemafibrate for 12 weeks. Like fenofibrate, pemafibrate significantly suppressed HFD-induced body weight gain; decreased plasma glucose, insulin and triglyceride (TG) levels; and increased plasma fibroblast growth factor 21 (FGF21). However, compared to the dose of fenofibrate, a relatively low dose of pemafibrate showed these effects. Pemafibrate activated PPARα transcriptional activity in the liver, increasing both hepatic expression and plasma levels of FGF21. Additionally, pemafibrate increased the expression of genes involved in thermogenesis and fatty acid oxidation, including Ucp1, Cidea and Cpt1b in inguinal adipose tissue (iWAT) and the mitochondrial marker Elovl3 in brown adipose tissue (BAT). Therefore, pemafibrate activates thermogenesis in iWAT and BAT by increasing plasma levels of FGF21. Additionally, pemafibrate induced the expression of Atgl and Hsl in epididymal white adipose tissue, leading to the activation of lipolysis. Taken together, pemafibrate suppresses diet-induced obesity in mice and improves their obesity-related metabolic abnormalities. We propose that pemafibrate may be useful for the suppression and improvement of obesity-induced metabolic abnormalities.


Subject(s)
Anti-Obesity Agents/therapeutic use , Benzoxazoles/therapeutic use , Butyrates/therapeutic use , Obesity/drug therapy , PPAR alpha/antagonists & inhibitors , Acetyltransferases/genetics , Acetyltransferases/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Benzoxazoles/administration & dosage , Benzoxazoles/pharmacology , Blood Glucose/metabolism , Butyrates/administration & dosage , Butyrates/pharmacology , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Diet, High-Fat/adverse effects , Fatty Acid Elongases , Insulin/blood , Lipolysis , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/prevention & control , Triglycerides/blood , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
14.
Biochem Biophys Res Commun ; 493(1): 40-45, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28928093

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitors have both anti-diabetic and anti-obesity effects. However, the precise mechanism of the anti-obesity effect remains unclear. We previously demonstrated that the glycogen depletion signal triggers lipolysis in adipose tissue via liver-brain-adipose neurocircuitry. In this study, therefore, we investigated whether the anti-obesity mechanism of SGLT2 inhibitor is mediated by this mechanism. Diet-induced obese mice were subjected to hepatic vagotomy (HVx) or sham operation and loaded with high fat diet containing 0.015% tofogliflozin (TOFO), a highly selective SGLT2 inhibitor, for 3 weeks. TOFO-treated mice showed a decrease in fat mass and the effect of TOFO was attenuated in HVx group. Although both HVx and sham mice showed a similar level of reduction in hepatic glycogen by TOFO treatment, HVx mice exhibited an attenuated response in protein phosphorylation by protein kinase A (PKA) in white adipose tissue compared with the sham group. As PKA pathway is known to act as an effector of the liver-brain-adipose axis and activate triglyceride lipases in adipocytes, these results indicated that SGLT2 inhibition triggered glycogen depletion signal and actuated liver-brain-adipose axis, resulting in PKA activation in adipocytes. Taken together, it was concluded that the effect of SGLT2 inhibition on weight loss is in part mediated via the liver-brain-adipose neurocircuitry.


Subject(s)
Adipose Tissue/physiology , Benzhydryl Compounds/administration & dosage , Brain/physiology , Glucosides/administration & dosage , Liver/physiology , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2/metabolism , Weight Loss/physiology , Adipose Tissue/drug effects , Adipose Tissue/innervation , Animals , Anti-Obesity Agents/administration & dosage , Brain/drug effects , Liver/drug effects , Liver/innervation , Male , Mice , Mice, Inbred C57BL , Vagotomy , Vagus Nerve/drug effects , Vagus Nerve/physiology , Vagus Nerve/surgery
15.
J Pharmacol Sci ; 133(4): 214-222, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28366492

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) is a well-known therapeutic target for treating hyperlipidemia. K-877 is a novel selective PPARα modulator (SPPARMα) that enhances PPARα transcriptional activity with high selectivity and potency, resulting in reduced plasma lipid levels. This study aimed to evaluate the effects of K-877 on hyperlipidemia in low-density lipoprotein receptor knockout (Ldlr-/-) mice, a mouse model of atherosclerosis. We revealed that K-877 administration significantly decreased plasma triglyceride (TG) and total cholesterol (TC) levels and increased plasma high-density lipoprotein cholesterol (HDL-C) levels in Ldlr-/- mice. K-877 administration to Ldlr-/- mice efficiently increased the gene expression of PPARα and its target genes related to fatty acid oxidation in the liver and small intestine. The same treatment significantly increased ATP-binding cassette a1 gene expression in the liver and small intestine and reduced Niemann Pick C1-like 1 gene expression in the small intestine, suggesting that K-877 administration induced HDL-C production in the liver and small intestine and reduced cholesterol absorption in the small intestine. In conclusion, K-877 administration had pronounced effects on the liver and small intestine in Ldlr-/- mice. K-877 is an attractive PPARα-modulating drug for treating hyperlipidemia that works equally well in both the liver and small intestine.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/genetics , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Butyrates/pharmacology , Butyrates/therapeutic use , Gene Expression/drug effects , Hyperlipidemias/drug therapy , Hyperlipidemias/genetics , Intestine, Small/metabolism , Lipid Metabolism/drug effects , PPAR alpha/agonists , PPAR alpha/genetics , Receptors, LDL/genetics , Animals , Atherosclerosis/metabolism , Cholesterol/metabolism , Disease Models, Animal , Fatty Acids/metabolism , Gene Knockout Techniques , Hyperlipidemias/metabolism , Intestinal Absorption/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Oxidation-Reduction/drug effects
16.
Biochem Biophys Res Commun ; 466(3): 536-40, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26381177

ABSTRACT

HMG-CoA reductase (HMGCR) catalyzes the conversion of HMG-CoA to mevalonic acid (MVA); this is the rate-limiting enzyme of the mevalonate pathway that synthesizes cholesterol. Statins, HMGCR inhibitors, are widely used as cholesterol-reducing drugs. However, statin-induced myopathy is the most adverse side effect of statins. To eludicate the mechanisms underlying statin the myotoxicity and HMGCR function in the skeletal muscle, we developed the skeletal muscle-specific HMGCR knockout mice. Knockout mice exhibited postnatal myopathy with elevated serum creatine kinase levels and necrosis. Myopathy in knockout mice was completely rescued by the oral administration of MVA. These results suggest that skeletal muscle toxicity caused by statins is dependent on the deficiencies of HMGCR enzyme activity and downstream metabolites of the mevalonate pathway in skeletal muscles rather than the liver or other organs.


Subject(s)
Hydroxymethylglutaryl CoA Reductases/deficiency , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal/enzymology , Rhabdomyolysis/enzymology , Rhabdomyolysis/etiology , Animals , Cholesterol/metabolism , Creatine Kinase/blood , Disease Models, Animal , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Male , Mevalonic Acid/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscular Diseases/chemically induced , Muscular Diseases/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Biochem Biophys Res Commun ; 465(4): 857-63, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26321664

ABSTRACT

Fatty acid elongase 5 (ELOVL5) is an enzyme involved in the synthesis of polyunsaturated fatty acids. Sterol Regulatory Element-binding Protein (SREBP)-1 activates ELOVL5 and increases polyunsaturated fatty acid synthesis, which in turn negatively affects SREBP-1 expression. Thus, ELOVL5 has been established as an SREBP-1 target gene and an important component of the negative feedback loop of de novo lipogenesis. However, the human ELOVL5 promoter/enhancer has not been fully analyzed and the location of SREBP biding sites around the ELOVL5 gene has yet to be defined. Here we performed a detailed promoter/enhancer analysis of human ELOVL5 gene, and identified two new SREBP binding sites, one in the 10 kb upstream region and one in the exon 1. These two SRE motifs are conserved among mammals and the mechanism found in the present study by which SREBP activates ELOVL5 is considered to be common in mammals. Through these findings, we clarified the molecular mechanism how SREBP activates ELOVL5, an important regulator of de novo lipogenesis.


Subject(s)
Acetyltransferases/genetics , Enhancer Elements, Genetic , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , Base Sequence , Binding Sites/genetics , Exons , Fatty Acid Elongases , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , HEK293 Cells , Humans , Lipogenesis/genetics , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Up-Regulation
18.
Hepatology ; 59(5): 1791-802, 2014 May.
Article in English | MEDLINE | ID: mdl-24277692

ABSTRACT

UNLABELLED: Liver X receptor (LXR) activation stimulates triglyceride (TG) accumulation in the liver. Several lines of evidence indicate that estradiol-17ß (E2) reduces TG levels in the liver; however, the molecular mechanism underlying the E2 effect remains unclear. Here, we show that administration of E2 attenuated sterol regulatory element-binding protein (SREBP)-1 expression and TG accumulation induced by LXR activation in mouse liver. In estrogen receptor alpha (ERα) knockout (KO) and liver-specific ERα KO mice, E2 did not affect SREBP-1 expression or TG levels. Molecular analysis revealed that ERα is recruited to the SREBP-1c promoter through direct binding to LXR and inhibits coactivator recruitment to LXR in an E2-dependent manner. Our findings demonstrate the existence of a novel liver-dependent mechanism controlling TG accumulation through the nonclassical ER/LXR pathway. To confirm that a nonclassical ER/LXR pathway regulates ERα-dependent inhibition of LXR activation, we screened ERα ligands that were able to repress LXR activation without enhancing ERα transcriptional activity, and, as a result, we identified the phytoestrogen, phloretin. In mice, phloretin showed no estrogenic activity; however, it did reduce SREBP-1 expression and TG levels in liver of mice fed a high-fat diet to an extent similar to that of E2. CONCLUSION: We propose that ER ligands reduce TG levels in the liver by inhibiting LXR activation through a nonclassical pathway. Our results also indicate that the effects of ER on TG accumulation can be distinguished from its estrogenic effects by a specific ER ligand.


Subject(s)
Fatty Liver/prevention & control , Orphan Nuclear Receptors/physiology , Receptors, Estrogen/physiology , Animals , Diet, High-Fat , Estradiol/pharmacology , Female , Ligands , Liver X Receptors , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/antagonists & inhibitors , Phloretin/pharmacology , Promoter Regions, Genetic , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics , Transcriptional Activation , Triglycerides/metabolism
19.
Biochem Biophys Res Commun ; 450(1): 318-23, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24938128

ABSTRACT

ELOVL family member 6, elongation of very long-chain fatty acids (Elovl6) is a microsomal enzyme that regulates the elongation of C12-16 saturated and monounsaturated fatty acids and is related to the development of obesity-induced insulin resistance via the modification of the fatty acid composition. In this study, we investigated the role of systemic Elovl6 in the pancreatic islet and ß-cell function. Elovl6 is expressed in both islets and ß-cell lines. In mice fed with chow, islets of the Elovl6(-/-) mice displayed normal architecture and ß-cell mass compared with those of the wild-type mice. However, when fed a high-fat, high-sucrose (HFHS) diet, the islet hypertrophy in response to insulin resistance observed in normal mice was attenuated and glucose-stimulated insulin secretion (GSIS) increased in the islets of Elovl6(-/-) mice compared with those of the wild-type mice. Enhanced GSIS in the HFHS Elovl6(-/-) islets was associated with an increased ATP/ADP ratio and the suppression of ATF-3 expression. Our findings suggest that Elovl6 could be involved in insulin secretory capacity per ß-cell and diabetes.


Subject(s)
Acetyltransferases/metabolism , Dietary Fats/adverse effects , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Obesity/metabolism , Obesity/pathology , Animals , Cells, Cultured , Fatty Acid Elongases , Female , Insulin Resistance , Insulin Secretion , Male , Mice , Mice, Knockout , Obesity/etiology , Organ Specificity , Tissue Distribution
20.
Nat Med ; 13(10): 1193-202, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17906635

ABSTRACT

Insulin resistance is often associated with obesity and can precipitate type 2 diabetes. To date, most known approaches that improve insulin resistance must be preceded by the amelioration of obesity and hepatosteatosis. Here, we show that this provision is not mandatory; insulin resistance and hyperglycemia are improved by the modification of hepatic fatty acid composition, even in the presence of persistent obesity and hepatosteatosis. Mice deficient for Elovl6, the gene encoding the elongase that catalyzes the conversion of palmitate to stearate, were generated and shown to become obese and develop hepatosteatosis when fed a high-fat diet or mated to leptin-deficient ob/ob mice. However, they showed marked protection from hyperinsulinemia, hyperglycemia and hyperleptinemia. Amelioration of insulin resistance was associated with restoration of hepatic insulin receptor substrate-2 and suppression of hepatic protein kinase C epsilon activity resulting in restoration of Akt phosphorylation. Collectively, these data show that hepatic fatty acid composition is a new determinant for insulin sensitivity that acts independently of cellular energy balance and stress. Inhibition of this elongase could be a new therapeutic approach for ameliorating insulin resistance, diabetes and cardiovascular risks, even in the presence of a continuing state of obesity.


Subject(s)
Acetyltransferases/metabolism , Diet, Atherogenic , Dietary Fats/pharmacology , Insulin Resistance , Obesity/metabolism , Acetyltransferases/deficiency , Acetyltransferases/genetics , Animals , Body Weight/drug effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Dietary Fats/administration & dosage , Fatty Acid Elongases , Gene Deletion , Insulin/metabolism , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/physiology , Liver Neoplasms/pathology , Male , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/genetics , Phosphoproteins/physiology , Phosphorylation , Protein Kinase C-epsilon/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL