Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 14(12): 1807-1814, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38116427

ABSTRACT

(+)-Negamycin, which is a dipeptide-like antibiotic containing a hydrazide structure, exhibits readthrough activity, resulting in the restoration of dystrophin in the mdx mouse model of Duchenne muscular dystrophy (DMD). In our previous structure-activity relationship study of negamycin, we found that its natural analogue 3-epi-deoxynegamycin (TCP-107), without antimicrobial activity, showed a higher readthrough activity than negamycin. In this study, we designed and synthesized cyclopropane-based conformationally restricted derivatives of TCP-107 and evaluated their readthrough activity in the cell-based reporter assay against a TGA-type mutation derived from DMD. As a result, a down-cis isomer, TCP-304, showed significant readthrough activity among the four isomers. Moreover, TCP-306, a derivative acylated by l-α-aminoundecanoic acid, possessed approximately 3 times higher activity than TCP-304. These down-cis derivatives showed dose-dependent readthrough activity and were effective for not only TGA but also TAG mutations. These results suggest that the conformational restriction of negamycin derivatives by the introduction of the cyclopropane ring is effective for an exhibition of potent readthrough activity.

2.
PNAS Nexus ; 2(11): pgad351, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37954160

ABSTRACT

The endoplasmic reticulum (ER)-embedded transcription factors, sterol regulatory element-binding proteins (SREBPs), master regulators of lipid biosynthesis, are transported to the Golgi for proteolytic activation to tune cellular cholesterol levels and regulate lipogenesis. However, mechanisms by which the cell responds to the levels of saturated or unsaturated fatty acids remain underexplored. Here, we show that RHBDL4/RHBDD1, a rhomboid family protease, directly cleaves SREBP-1c at the ER. The p97/VCP, AAA-ATPase complex then acts as an auxiliary segregase to extract the remaining ER-embedded fragment of SREBP-1c. Importantly, the enzymatic activity of RHBDL4 is enhanced by saturated fatty acids (SFAs) but inhibited by polyunsaturated fatty acids (PUFAs). Genetic deletion of RHBDL4 in mice fed on a Western diet enriched in SFAs and cholesterol prevented SREBP-1c from inducing genes for lipogenesis, particularly for synthesis and incorporation of PUFAs, and secretion of lipoproteins. The RHBDL4-SREBP-1c pathway reveals a regulatory system for monitoring fatty acid composition and maintaining cellular lipid homeostasis.

3.
Eur J Obstet Gynecol Reprod Biol ; 214: 65-70, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28482330

ABSTRACT

OBJECTIVE: C-C motif chemokine ligand 20 is thought to contribute to the development of endometriosis by recruiting Th17 lymphocytes into endometriotic foci. The present study investigated the effects of dienogest, a progesterone receptor agonist used to treat endometriosis, on C-C motif chemokine ligand 20 expression by endometriotic cells. STUDY DESIGN: Effects of dienogest on mRNA expression and protein secretion of C-C motif chemokine ligand 20 induced by interleukin 1ß were assessed in three immortalized endometriotic epithelial cell lines, parental cells (EMosis-CC/TERT1), and stably expressing human progesterone receptor isoform A (EMosis-CC/TERT1/PRA+) or isoform B (EMosis-CC/TERT1/PRA-/PRB+). RESULTS: Dienogest markedly inhibited interleukin 1ß-stimulated C-C motif chemokine ligand 20 mRNA expression and protein secretion in EMosis-CC/TERT1/PRA-/PRB+, which was abrogated by the progesterone receptor antagonist RU486. In EMosis-CC/TERT1/PRA+, dienogest slightly inhibited C-C motif chemokine ligand 20 mRNA and protein. In EMosis-CC/TERT1, dienogest slightly inhibited C-C motif chemokine ligand 20 mRNA, but had no effect on C-C motif chemokine ligand 20 protein. CONCLUSION: Dienogest inhibited interleukin 1ß-induced up-regulation of C-C motif chemokine ligand 20 in endometriotic epithelial cells, mainly mediated by progesterone receptor B.


Subject(s)
Chemokine CCL20/metabolism , Endometriosis/drug therapy , Epithelial Cells/drug effects , Nandrolone/analogs & derivatives , Receptors, Progesterone/agonists , Cell Line , Epithelial Cells/metabolism , Female , Humans , Interleukin-1beta , Mifepristone , Nandrolone/pharmacology , Nandrolone/therapeutic use , Receptors, Progesterone/metabolism
4.
J Atheroscler Thromb ; 22(11): 1172-85, 2015.
Article in English | MEDLINE | ID: mdl-26016511

ABSTRACT

AIM: Vascular endothelial dysfunction is considered an early predictor of atherosclerosis. It has been proven that elevated blood levels of free fatty acids pose a substantial risk for the development of cardiovascular disease. In this study, we examined the effects of palmitic acid (PA), a saturated fatty acid, on endothelial function by using the expression of adhesion molecule, cytokines, and inflammatory protein as indicators, as well as investigated the effects of eicosapentaenoic acid, an n-3 polyunsaturated fatty acid. METHODS: Human umbilical vein endothelial cells (HUVEC) were exposed to PA and EPA. RESULTS: When HUVEC were exposed to PA, there was an increase in the expression of adhesion molecule, cytokines, and inflammatory protein (ICAM-1, MCP-1, interleukin-6, PTX3). PA augmented the expression of long-chain acyl-CoA synthetase (ACSL) and the cyclin-dependent kinase inhibitor p21, and enhanced the phosphorylation of p65, a component of NF-κB. ACSL inhibition and siRNA-mediated ACSL3 knockdown suppressed the PA-induced increase in the expression of adhesion molecule, cytokines, and inflammatory protein, and ACSL inhibition suppressed the enhancement of p65 phosphorylation. In addition, p21 knockdown suppressed the PA-induced increase in the expression of MCP-1 and ICAM-1. EPA suppressed the PA-induced increase in the expression of ACSL and p21, the enhancement of p65 phosphorylation, as well as the associated increase in the expression of ICAM-1, MCP-1, interleukin-6, and PTX3. CONCLUSIONS: These results suggest that the ACSL, p21, and NF-κB-dependent pathway may possibly be involved in PA-induced vascular endothelial dysfunction, and that EPA ameliorates this at least in part through the regulation of ACSL3 expression.


Subject(s)
Coenzyme A Ligases/metabolism , Eicosapentaenoic Acid/pharmacology , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Palmitic Acids/adverse effects , Blotting, Western , Cells, Cultured , Cytokines/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Phosphorylation/drug effects
5.
Sci Rep ; 4: 6406, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25230773

ABSTRACT

Small-vessel vasculitis is a life-threatening autoimmune disease that is frequently associated with anti-neutrophil cytoplasmic antibodies (ANCAs). Conventional immunotherapy including steroids and cyclophosphamide can cause serious adverse events, limiting the efficacy and safety of treatment. Eicosapentaenoic acid (EPA), a key component of fish oil, is an omega-3 polyunsaturated fatty acid widely known to be cardioprotective and beneficial for vascular function. We report two elderly patients with systemic ANCA-associated vasculitis (AAV) in whom the administration of EPA in concert with steroids safely induced and maintained remission, without the use of additioal immunosuppressants. To explore the mechanisms by which EPA enhances the treatment of AAV, we employed SCG/Kj mice as a spontaneous murine model of AAV. Dietary enrichment with EPA significantly delayed the onset of crescentic glomerulonephritis and prolonged the overall survival. EPA-derived anti-inflammatory lipid mediators and their precursors were present in the kidney, plasma, spleen, and lungs in the EPA-treated mice. Furthermore, a decrease in ANCA production and CD4/CD8-double negative T cells, and an increase in Foxp3(+) regulatory T cells in the lymph nodes of the kidney were observed in the EPA-treated mice. These clinical and experimental observations suggest that EPA can safely support and augment conventional therapy for treating autoimmune small-vessel vasculitis.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/drug therapy , Disease Models, Animal , Eicosapentaenoic Acid/therapeutic use , Immunomodulation/drug effects , Aged , Animals , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/pathology , Blotting, Western , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Proliferation , Cells, Cultured , Female , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred Strains , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
6.
J Biochem ; 155(5): 301-13, 2014 May.
Article in English | MEDLINE | ID: mdl-24729033

ABSTRACT

The proteolytic cascade is the key step in transactivation of sterol regulatory element-binding proteins (SREBPs), a transcriptional factor of lipid synthesis. Proteolysis of SREBP-2 is strictly regulated by sterols, but that of SREBP-1c was not strongly sterol-regulated, but inhibited by polyunsaturated fatty acids (PUFAs). In this study, the proteolytic processing of SREBP-1 and -2 was examined by transfection studies of cDNA-encoding mutants in which all the known cleavage sites were disrupted. In cultured cells, sterol-regulated SREBP-2 processing was completely eliminated by mutation of cleavage sites. In contrast, the corresponding SREBP-1c mutants as well as wild type exhibited large amounts of cleaved products in the nuclear extracts from culture cells and murine liver in vivo. The nuclear form of the mutant SREBP-1c was induced by delipidated condition and suppressed by eicosapentaenoic acid, an n-3 PUFA, but not by sterols. This novel processing mechanism was affected by neither SREBP cleavage-activating protein (SCAP) nor insulin-induced gene (Insig)-1, unlike SREBP-2, but abolished by a serine protease inhibitor. Through analysis of deletion mutant, a site-2 protease recognition sequence (DRSR) was identified to be involved in this novel processing. These findings suggest that SREBP-1c cleavage could be subjected to a novel PUFA-regulated cleavage system in addition to the sterol-regulatory SCAP/Insig system.


Subject(s)
Fatty Acids, Unsaturated/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Fatty Acids, Unsaturated/pharmacology , HEK293 Cells/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Protein Processing, Post-Translational , Rats , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Sterols/metabolism , Sterols/pharmacology
7.
Article in English | MEDLINE | ID: mdl-23375839

ABSTRACT

Despite established anti-atherogenic action, previous reports have shown that fish oils or n-3 poly-unsaturated fatty acid (PUFA) increase plasma LDL-C in animals and humans. However, which component of n-3 PUFAs and what mechanisms contribute to this increase are unclear. We investigated the effects of the major components of n-3 PUFA, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), on plasma LDL-C in high fat diet-fed hamsters. While LDL-C increased significantly with n-3 PUFA oil and DHA, EPA had no effect on LDL-C. Interestingly, a positive correlation was found between plasma cholesterol ester transfer protein (CETP) activity and LDL-C. Only DHA increased plasma CETP activity and significantly decreased LDL receptor expression in the liver. Our data suggest that DHA, not EPA, is a major factor in the LDL-C increasing effect of n-3 PUFA oil. These differential effects on LDL-C may arise from differences in plasma CETP activity and LDL receptor expression.


Subject(s)
Cholesterol Ester Transfer Proteins/metabolism , Cholesterol, LDL/blood , Diet, High-Fat/adverse effects , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Receptors, LDL/metabolism , Animals , Cricetinae , Fatty Acids, Omega-3/pharmacology
8.
Atherosclerosis ; 227(2): 289-96, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23433401

ABSTRACT

BACKGROUND: Chronic inflammation caused by macrophages may be associated with progression of arteriosclerosis or obesity, both risk factors for cardiovascular events. In the Japan EPA Lipid Intervention Study (JELIS), eicosapentaenoic acid (EPA), an n-3 polyunsaturated fatty acid, was found to reduce the incidence of cardiovascular events. METHODS: The effect of EPA on the expression of inflammatory factors induced by palmitate, a saturated fatty acid, was investigated using human THP-1 macrophages. RESULTS: Palmitate induced expression of inflammatory cytokines and activated NF-κB, similar to lipopolysaccharide (LPS). EPA strongly suppressed palmitate-induced up-regulation of inflammatory factors while slightly suppressing LPS-induced factors. Both palmitate and LPS up-regulated expression of long-chain acyl-CoA synthetase (ACSL) 1, while EPA preferentially suppressed palmitate-induced ACSL1 expression. Although an acyl-CoA synthetase inhibitor and ACSL1 siRNA both suppressed palmitate-induced tumor necrosis factor (TNF)-α expression, the former had no effect on LPS-induced TNF-α expression. Palmitate may therefore stimulate cytokine production through a different mechanism than LPS mediated through Toll-like receptor 4, at least partly, and ACSL1 may play an important role in this mechanism. Finally, palmitate induced expression of sterol regulatory element-binding protein-1a and ACSL1, while EPA suppressed the expression of these genes. CONCLUSION: The suppressive effects of EPA on palmitate-induced cytokine production may be mediated by the suppression of ACSL1 expression, at least partly. This anti-inflammatory effect of EPA may contribute to suppression of chronic inflammation caused by macrophages in atherosclerotic plaques.


Subject(s)
Coenzyme A Ligases/biosynthesis , Coenzyme A Ligases/metabolism , Cytokines/metabolism , Eicosapentaenoic Acid/metabolism , Gene Expression Regulation, Enzymologic , Macrophages/enzymology , Palmitic Acid/metabolism , Cell Line , Coenzyme A Ligases/chemistry , Humans , Inflammation , Japan , Macrophages/cytology , Sterol Regulatory Element Binding Protein 1/metabolism , Toll-Like Receptor 4/metabolism
9.
Atherosclerosis ; 219(2): 566-72, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22018640

ABSTRACT

High level of plasma very low-density lipoprotein (VLDL) has been identified as a risk factor for coronary heart disease. Recent evidence suggests that excess VLDL induces inflammatory responses in macrophages and vascular endothelial cells. The Japan EPA Lipid Intervention Study (JELIS), a large scale clinical trial, demonstrated that highly purified eicosapentaenoic acid (EPA) prevented the onset of cardiovascular events in LDL-cholesterol independent fashion. In this study, we investigated the impact of EPA on pro-inflammatory properties of VLDL. Effects of VLDL prepared from mice fed 5% EPA diet for 1 week (EPA-VLDL) or mice fed normal diet (Ctrl-VLDL) on the mRNA expression of pro-inflammatory factors were examined in human THP-1 macrophages. Ctrl-VLDL increased mRNA expression of pro-inflammatory factors such as interleukin-1ß and tumor necrosis factor-α in macrophages. In contrast, the increases in pro-inflammatory factors by EPA-VLDL were lower than those by Ctrl-VLDL. Moreover, EPA-VLDL-treated macrophages had less triglyceride accumulation than Ctrl-VLDL-treated macrophages. Inhibition of lipoprotein lipase (LPL) appeared to suppress inflammation and triglyceride accumulation by Ctrl-VLDL suggesting that hydrolysis of VLDL is required for the pro-inflammatory properties of VLDL. Free fatty acid release from EPA-VLDL by macrophages and purified LPL was less than that from Ctrl-VLDL. Extracellular LPL mass was decreased by EPA-VLDL. Taken together, these findings indicate that the pro-inflammatory properties of VLDL were attenuated by EPA administration via decrease in susceptibility of VLDL to LPL. It appears possible that anti-inflammatory effects of EPA on VLDL contribute to the suppression of cardiovascular risk by EPA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Eicosapentaenoic Acid/pharmacology , Lipoprotein Lipase/metabolism , Lipoproteins, VLDL/metabolism , Macrophages/drug effects , Animals , Apolipoprotein C-II/blood , Apolipoprotein C-III/blood , Biomarkers/blood , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrolysis , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lactones/pharmacology , Lipoprotein Lipase/antagonists & inhibitors , Lipoproteins, VLDL/blood , Macrophages/enzymology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Orlistat , Particle Size , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
J Biol Chem ; 286(47): 40835-46, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21911492

ABSTRACT

F-box and WD repeat domain-containing 7α (Fbw7α) is the substrate recognition component of a ubiquitin ligase that controls the degradation of factors involved in cellular growth, including c-Myc, cyclin E, and c-Jun. In addition, Fbw7α degrades the nuclear form of sterol regulatory element-binding protein (SREBP)-1a, a global regulator of lipid synthesis, particularly during mitosis in cultured cells. This study investigated the in vivo role of Fbw7α in hepatic lipid metabolism. siRNA knockdown of Fbw7α in mice caused marked hepatosteatosis with the accumulation of triglycerides. However, inhibition of Fbw7α did not change the level of nuclear SREBP-1 protein or the expression of genes involved in fatty acid synthesis and oxidation. In vivo experiments on the gain and loss of Fbw7α function indicated that Fbw7α regulated the expression of peroxisome proliferator-activated receptor (PPAR) γ2 and its target genes involved in fatty acid uptake and triglyceride synthesis. These genes included fatty acid transporter Cd36, diacylglycerol acyltransferase 1 (Dgat1), and fat-specific protein 27 (Cidec). The regulation of PPARγ2 by Fbw7α was mediated, at least in part, by the direct degradation of the Krüppel-like factor 5 (KLF5) protein, upstream of PPARγ2 expression. Hepatic Fbw7α contributes to normal fatty acid and triglyceride metabolism, functions that represent novel aspects of this cell growth regulator.


Subject(s)
F-Box Proteins/metabolism , Fatty Liver/metabolism , Kruppel-Like Transcription Factors/metabolism , PPAR gamma/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Fatty Liver/genetics , Fatty Liver/pathology , Fenofibrate/pharmacology , Gene Knockdown Techniques , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , PPAR gamma/deficiency , PPAR gamma/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
11.
Eur J Pharmacol ; 669(1-3): 128-35, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21871881

ABSTRACT

Fish oil containing n-3 polyunsaturated fatty acids such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) is well known to prevent the progression of IgA nephropathy. However, the mechanism through which fish oil prevents the progression of renal injury remains uncertain. We tried to clarify the effects of EPA on tubulointerstitial injury in the kidney both in vivo and in vitro. We examined the effects of EPA, especially to focus on nuclear factor kappa B (NF-κB), using Thy-1 nephritis models. Also the mechanism of EPA was investigated using small-interfering RNA (siRNA) in lipopolysaccharide (LPS)-stimulated proximal tubular epithelial cells (PTECs). In Thy-1 nephritis models, EPA significantly inhibited tubulointerstitial injury and the infiltration of macrophages into tubulointerstitial lesions except severe glomerular injury at early stage. Compared with control animals, NF-κB activation was significantly augmented in the Thy-1 nephritic kidney. However, treatment with EPA significantly reduced NF-κB activation, down-regulated the expressions of NF-κB-dependent molecules. Also in LPS-stimulated PTECs, LPS augmented NF-κB activation and the expression of NF-κB-dependent molecules. As in the case with the Thy-1 nephritis models, treatment with EPA inhibited them, prevented the degradation of IκBα in LPS-stimulated PTECs. Pre-treatment with siRNA for IκBα abolished the inhibitory effect of EPA on LPS-induced NF-κB activation, suggesting that EPA inhibited NF-κB activation by regulating IκBα. Our results indicate that EPA prevents the early progression of tubulointerstitial injury in Thy-1 nephritis models, and the inhibitory effect of EPA on the expression of inflammatory molecules via the regulation of IκBα in cultured cells may explain this mechanism.


Subject(s)
Eicosapentaenoic Acid/pharmacology , I-kappa B Proteins/metabolism , NF-kappa B/antagonists & inhibitors , Nephritis, Interstitial/drug therapy , Protective Agents/pharmacology , Proteinuria/drug therapy , Animals , Cells, Cultured , Epithelial Cells/metabolism , Female , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/antagonists & inhibitors , Kidney Tubules, Proximal/cytology , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha , Nephritis, Interstitial/metabolism , Proteinuria/metabolism , Rats , Rats, Wistar , Vascular Cell Adhesion Molecule-1/metabolism
12.
Diabetes ; 59(10): 2495-504, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20682690

ABSTRACT

OBJECTIVE: Given the pleiotropic effect of eicosapentaenoic acid (EPA), it is interesting to know whether EPA is capable of improving obesity. Here we examined the anti-obesity effect of EPA in mice with two distinct models of obesity. RESEARCH DESIGN AND METHODS: Male C57BL/6J mice were fed a high-fat/high-sucrose diet (25.0% [w/w] fat, 32.5% [w/w] sucrose) (HF/HS group) or a high-fat diet (38.1% [w/w] fat, 8.5% [w/w] sucrose) (HF group) for 4-20 weeks. A total of 5% EPA was administered by partially substituting EPA for fat in the HF/HS + EPA and HF + EPA groups. RESULTS: Both the HF/HS and HF groups similarly developed obesity. EPA treatment strongly suppresses body weight gain and obesity-related hyperglycemia and hyperinsulinemia in HF/HS-fed mice (HF/HS + EPA group), where hepatic triglyceride content and lipogenic enzymes are increased. There is no appreciable effect of EPA on body weight in HF-fed mice (HF + EPA group) without enhanced expression of hepatic lipogenic enzymes. Moreover, EPA is capable of reducing hepatic triglyceride secretion and changing VLDL fatty acid composition in the HF/HS group. By indirect calorimetry analysis, we also found that EPA is capable of increasing energy consumption in the HF/HS + EPA group. CONCLUSIONS: This study is the first demonstration that the anti-obesity effect of EPA in HF/HS-induced obesity is associated with the suppression of hepatic lipogenesis and steatosis. Because the metabolic syndrome is often associated with hepatic lipogenesis and steatosis, the data suggest that EPA is suited for treatment of the metabolic syndrome.


Subject(s)
Anti-Obesity Agents/pharmacology , Dietary Fats/pharmacology , Dietary Sucrose/pharmacology , Eicosapentaenoic Acid/pharmacology , Animals , Blood Glucose/metabolism , Cholesterol/blood , Coronary Disease/prevention & control , Energy Intake , Fatty Acids, Nonesterified/blood , Fish Oils/therapeutic use , Humans , Hyperglycemia/chemically induced , Hyperglycemia/prevention & control , Hyperinsulinism/chemically induced , Hyperinsulinism/prevention & control , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/prevention & control , Triglycerides/blood , Weight Gain/drug effects
13.
Article in English | MEDLINE | ID: mdl-20542418

ABSTRACT

Cide-a and Cide-c belong to the cell death-inducing DNA fragmentation factor-alpha-like effector family. Recent evidences suggest that these proteins may be involved in lipid accumulation in liver and adipose tissues. We confirmed that in the high-fat/high-sucrose diet-induced murine model of hepatic steatosis, the expression levels of the Cide-a and Cide-c genes were markedly and time-dependently increased, but returned to normal levels following improvement of hepatic steatosis by eicosapentaenoic acid (EPA) administration. Levels of expression of the Cide-a and Cide-c genes correlated well with plasma ALT. EPA inhibited the promoter activity of the Cide-a gene in vitro. Sterol regulatory element-binding protein-1 (SREBP-1) markedly enhanced the promoter activity of Cide-a, and EPA inhibited the expression of Cide-a mRNA. SREBP-1 and EPA did not affect those of Cide-c. These findings indicate that Cide-a and Cide-c are closely involved in the progression of hepatic steatosis, and that EPA inhibits Cide-a gene expression through SREBP-1 regulation.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Eicosapentaenoic Acid/pharmacology , Fatty Liver/drug therapy , Fatty Liver/metabolism , Gene Expression Regulation/drug effects , Proteins/metabolism , Adipose Tissue/metabolism , Animals , Diet/adverse effects , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Disease Models, Animal , Fatty Liver/chemically induced , Liver/metabolism , Male , Mice , Promoter Regions, Genetic , Sterol Regulatory Element Binding Protein 1/metabolism , Sucrose/adverse effects , Sucrose/pharmacology , Sweetening Agents/adverse effects , Sweetening Agents/pharmacology
14.
Diabetes ; 57(9): 2382-92, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18458149

ABSTRACT

OBJECTIVE: Chronic exposure to fatty acids causes beta-cell failure, often referred to as lipotoxicity. We investigated its mechanisms, focusing on contribution of SREBP-1c, a key transcription factor for lipogenesis. RESEARCH DESIGN AND METHODS: We studied in vitro and in vivo effects of saturated and polyunsaturated acids on insulin secretion, insulin signaling, and expression of genes involved in beta-cell functions. Pancreatic islets isolated from C57BL/6 control and SREBP-1-null mice and adenoviral gene delivery or knockdown systems of related genes were used. RESULTS: Incubation of C57BL/6 islets with palmitate caused inhibition of both glucose- and potassium-stimulated insulin secretion, but addition of eicosapentaenoate (EPA) restored both inhibitions. Concomitantly, palmitate activated and EPA abolished both mRNA and nuclear protein of SREBP-1c, accompanied by reciprocal changes of SREBP-1c target genes such as insulin receptor substrate-2 (IRS-2) and granuphilin. These palmitate-EPA effects on insulin secretion were abolished in SREBP-1-null islets. Suppression of IRS-2/Akt pathway could be a part of the downstream mechanism for the SREBP-1c-mediated insulin secretion defect because adenoviral constitutively active Akt compensated it. Uncoupling protein-2 (UCP-2) also plays a crucial role in the palmitate inhibition of insulin secretion, as confirmed by knockdown experiments, but SREBP-1c contribution to UCP-2 regulation was partial. The palmitate-EPA regulation of insulin secretion was similarly observed in islets from C57BL/6 mice pretreated with dietary manipulations. Furthermore, administration of EPA to diabetic KK-Ay mice ameliorated impairment of insulin secretion in their islets. CONCLUSIONS: SREBP-1c plays a dominant role in palmitate-mediated insulin secretion defect, and EPA prevents it through SREBP-1c inhibition, implicating a therapeutic potential for treating diabetes related to lipotoxicity.


Subject(s)
Eicosapentaenoic Acid/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Palmitates/toxicity , Sterol Regulatory Element Binding Protein 1/metabolism , Adenoviridae/genetics , Animals , Eicosapentaenoic Acid/metabolism , Gene Expression/drug effects , Gene Silencing , Insulin Receptor Substrate Proteins , Insulin Secretion , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Lipogenesis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Organ Culture Techniques , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 1/genetics , Uncoupling Protein 2
15.
FEBS J ; 274(17): 4440-52, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17662109

ABSTRACT

Sterol regulatory element-binding protein (SREBP)-1a is a unique membrane-bound transcription factor highly expressed in actively growing cells and involved in the biosynthesis of cholesterol, fatty acids, and phospholipids. Because mammalian cells need to synthesize membrane lipids for cell replication, the functional relevance of SREBP-1a in cell proliferation has been considered a biological adaptation. However, the effect of this potent lipid-synthesis activator on cell growth has never been explored. Here, we show that induction of nuclear SREBP-1a, but not SREBP-2, completely inhibited cell growth in inducible Chinese hamster ovary (CHO) cell lines. Growth inhibition occurred through G(1) cell-cycle arrest, which is observed in various cell types with transient expression of nuclear SREBP-1a. SREBP-1a caused the accumulation of cyclin-dependent kinase (cdk) inhibitors such as p27, p21, and p16, leading to reduced cdk2 and cdk4 activities and hypophosphorylation of Rb protein. In contrast to transactivation of p21, SREBP-1a activated p27 by enhancing stabilization of the protein through inhibition of SKP2 and KPC1. In vivo, SREBP-1a-expressing livers of transgenic mice exhibited impaired regeneration after partial hepatectomy. SREBP-1-null mouse embryonic fibroblasts had a higher cell proliferation rate than wild-type cells. The unexpected cell growth-inhibitory role of SREBP-1a provides a new paradigm to link lipid synthesis and cell growth.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , G1 Phase/physiology , Lipids/biosynthesis , Protein Kinase Inhibitors/pharmacology , Sterol Regulatory Element Binding Protein 1/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoprecipitation , Mice , Mice, Knockout , Mice, Transgenic , Sterol Regulatory Element Binding Protein 1/genetics
16.
Mol Cell Biol ; 25(20): 8938-47, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16199872

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) are membrane-bound transcription factors that regulate lipid synthetic genes. In contrast to SREBP-2, which regulates cellular cholesterol level in normal cells, SREBP-1a is highly expressed in actively growing cells and activates entire programs of genes involved in lipid synthesis such as cholesterol, fatty acids, triglycerides, and phospholipids. Previously, the physiological relevance of this potent activity of SREBP-1a has been thought to regulate the supply of membrane lipids in response to cell growth. Here we show that nuclear SREBP-1a and SREBP-2 bind directly to a novel SREBP binding site in the promoter of the p21(WAF1/CIP1) gene, the major cyclin-dependent kinase inhibitor, and strongly activate its promoter activity. Only the SREBP-1a isoform consistently causes induction of p21 at both the mRNA and protein levels. Colony formation assays and polyploidy of livers from transgenic mice suggest that activation of p21 by SREBP-1a could inhibit cell growth. Activation of endogenous SREBPs in lipid deprivation conditions was associated with induction of p21 mRNA and protein. Expression of p21 was reduced in SREBP-1 null mice. These data suggest a physiological role of SREBP-1a in p21 regulation. Identification of p21 as a new SREBP target might implicate a new paradigm in the link between lipid synthesis and cell growth.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Sterol Regulatory Element Binding Protein 1/pharmacology , Animals , Base Sequence , Cell Line , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA, Complementary/genetics , Enzyme Inhibitors/metabolism , Humans , In Vitro Techniques , Lipids/biosynthesis , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/pharmacology , Transcriptional Activation
17.
Nat Cell Biol ; 6(4): 351-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15048126

ABSTRACT

Insulin receptor substrate 2 (IRS-2) is the main mediator of insulin signalling in the liver, controlling insulin sensitivity. Sterol regulatory element binding proteins (SREBPs) have been established as transcriptional regulators of lipid synthesis. Here, we show that SREBPs directly repress transcription of IRS-2 and inhibit hepatic insulin signalling. The IRS-2 promoter is activated by forkhead proteins through an insulin response element (IRE). Nuclear SREBPs effectively replace and interfere in the binding of these transactivators, resulting in inhibition of the downstream PI(3)K/Akt pathway, followed by decreased glycogen synthesis. These data suggest a molecular mechanism for the physiological switching from glycogen synthesis to lipogenesis and hepatic insulin resistance that is associated with hepatosteatosis.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Liver/metabolism , Phosphoproteins/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Feedback, Physiological/genetics , Forkhead Transcription Factors , Glycogen/metabolism , Insulin Receptor Substrate Proteins , Insulin Resistance/genetics , Intracellular Signaling Peptides and Proteins , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Response Elements/physiology , Signal Transduction/genetics , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics
18.
Biochem Biophys Res Commun ; 316(3): 790-4, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-15033469

ABSTRACT

The ATP-binding-cassette transporter A1 (ABCA1) plays an essential role in cellular cholesterol efflux and helps prevent macrophages from becoming foam cells. The statins are widely used as cholesterol-lowering agents and have other anti-atherogenic actions. We tested the effects of four different statins (fluvastatin, atorvastatin, simvastatin, and lovastatin) on ABCA1 expression in macrophages in vitro. The statins suppressed ABCA1 mRNA expression in RAW246.7 and THP-1 macrophage cell lines and in mouse peritoneal macrophages. The effect was time- and dose-dependent and was abolished by the addition of the post-reductase product, mevalonate. These findings imply that there is a possible modulation of the well-known beneficial effects of the statins on the reverse cholesterol transport pathway.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/chemistry , Animals , Atorvastatin , Biological Transport , Blotting, Northern , Cell Line , Cholesterol/metabolism , Dose-Response Relationship, Drug , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Heptanoic Acids/pharmacology , Humans , Indoles/pharmacology , Lovastatin/pharmacology , Luciferases/metabolism , Macrophages/metabolism , Mice , Oxygen/metabolism , Promoter Regions, Genetic , Pyrroles/pharmacology , RNA, Messenger/metabolism , Simvastatin/pharmacology , Swiss 3T3 Cells , Time Factors , Transcription, Genetic
19.
J Biol Chem ; 279(13): 12027-35, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-14722127

ABSTRACT

The hepatocyte nuclear factor-4alpha (HNF-4alpha)/PGC-1 pathway plays a crucial role in the transcriptional regulation of hepatic gluconeogenic enzymes such as phosphoenolpyruvate carboxykinase (PEPCK) and Glc-6-Pase, genes that are activated at fasting and suppressed in a fed state. SREBP-1c dominates the nutritional regulation of lipogenic genes inverse to gluconeogenesis. Here we show the mechanism by which SREBP-1 suppresses expression of gluconeogenic genes. A series of luciferase reporter assays demonstrated that SREBP-1a and -1c effectively inhibited the PEPCK promoter activity that was induced by HNF-4alpha. The HNF-4alpha-binding site in the glucocorticoid-response unit was responsible for the SREBP-1 inhibition, although SREBP-1 did not bind to the PEPCK promoter as demonstrated by electrophoretic mobility shift assays. The inhibitory effect was more potent in the isoform of SREBP-1a than SREBP-1c and was eliminated by deletion of the amino-terminal transactivation domain of SREBP-1. Coimmunoprecipitation experiments demonstrated that these two transcription factors directly interact through the transactivation domain of SREBP-1 and the ligand binding/AF2 domains of HNF-4alpha. Estimation of coactivator recruitment using HNF-4alpha-Gal4DBD fusion assay showed that SREBP-1 competitively inhibited PGC-1 recruitment, a requirement for HNF-4alpha activation. Consistent with these results, hepatic PEPCK and Glc-6-Pase mRNA levels are suppressed by overexpression of SREBP-1a and -1c in the transgenic mice. Our data indicate that SREBP-1 has a novel role as negative regulator of gluconeogenic genes through a cross-talk with HNF-4alpha interference with PGC-1 recruitment.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Binding Sites , Binding, Competitive , Blotting, Northern , Cell Line , Colforsin/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation , Glutathione Transferase/metabolism , Hepatocyte Nuclear Factor 4 , Humans , Ligands , Lipoproteins, LDL/metabolism , Liver/metabolism , Luciferases/metabolism , Mice , Mice, Transgenic , Models, Genetic , Phosphoenolpyruvate Carboxykinase (GTP)/biosynthesis , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , RNA/metabolism , Recombinant Fusion Proteins/metabolism , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Transcription, Genetic , Transcriptional Activation , Transfection
20.
Hepatology ; 38(6): 1529-39, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14647064

ABSTRACT

Leptin-deficient ob/ob mice show many characteristics of obesity, including excess peripheral adiposity as well as severe hepatic steatosis, at least in part, due to increased hepatic lipogenesis. Polyunsaturated fatty acids (PUFAs) are not only ligands for peroxisome proliferator-activated receptor (PPAR) alpha but are also negative regulators of hepatic lipogenesis, which is thought to be mediated by the repression of sterol regulatory element-binding protein (SREBP)-1. We have previously shown that the disruption of SREBP-1 in ob/ob mice decreased their liver triglyceride storage. To examine whether PUFAs could reduce hepatic triglyceride deposition, we challenged ob/ob mice with dietary PUFA. It is demonstrated that PUFA markedly decreased the mature form of SREBP-1 protein and thereby reduced the expression of lipogenic genes such as fatty acid synthase (FAS) and stearoyl-CoA desaturase 1 (SCD1) in the livers of ob/ob mice. Consequently, the liver triglyceride content and plasma alanine aminotransferase (ALT) levels were decreased. Furthermore, both hyperglycemia and hyperinsulinemia in ob/ob mice were improved by PUFA administration, similar to the effect of PPARalpha activators. In conclusion, PUFAs ameliorate obesity-associated symptoms, such as hepatic steatosis and insulin resistance, presumably through both down-regulation of SREBP-1 and activation of PPARalpha.


Subject(s)
CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Fatty Acids, Unsaturated/pharmacology , Fatty Liver/drug therapy , Alanine Transaminase/blood , Animals , Cholesterol/biosynthesis , Fatty Acids, Unsaturated/therapeutic use , Gene Expression Regulation/drug effects , Insulin Resistance , Liver/chemistry , Mice , Mice, Inbred C57BL , Mice, Obese , Receptors, Cytoplasmic and Nuclear/physiology , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/physiology , Triglycerides/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...