Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 12(1): e0302423, 2024 01 11.
Article in English | MEDLINE | ID: mdl-38054720

ABSTRACT

ABSTRACT: In pathogenic bacteria, the flavohemoglobin Hmp is crucial in metabolizing the cytotoxic levels of nitric oxide (NO) produced in phagocytic cells, contributing to bacterial virulence. Hmp expression is predominantly regulated by the Rrf2 family transcription repressor NsrR in an NO-dependent manner; however, the underlying molecular mechanism in enterobacteria remains poorly understood. In this study, we identified Val43 of Salmonella Typhimurium NsrR (StNsrR) as a critical amino acid residue for regulating Hmp expression. The Val43-to-Ala-substituted mutant NsrR isolated through random and site-directed mutagenesis showed high binding affinity to the target DNA irrespective of NO exposure, resulting in a severe reduction in hmp transcription and slow NO metabolism in Salmonella under NO-producing conditions. Conversely, the Val43-to-Glu-substituted NsrR caused effects similar to nsrR null mutation, which directed hmp transcription and NO metabolism in a constitutive way. Comparative analysis of the primary sequences of NsrR and another NO-sensing Rrf2 family regulator, IscR, from diverse bacteria, revealed that Val43 of enterobacterial NsrR corresponds to Ala in Pseudomonas aeruginosa or Streptomyces coelicolor NsrR and Glu in enterobacterial IscR, all of which are located in the DNA recognition helix α3. The predicted structure of StNsrR in complex with the hmp DNA suggests dissimilar spatial stoichiometry in the interactions of Val43 and its substituted residues with the target DNA, consistent with the observed phenotypic changes in StNsrR Val43 mutants. Our findings highlight the discriminative roles of the NsrR recognition helix in regulating species-specific target gene expression, facilitating effective NO detoxification strategies in bacteria across diverse environments. IMPORTANCE: The precise regulation of flavohemoglobin Hmp expression by NsrR is critical for bacterial fitness, as excessive Hmp expression in the absence of NO can disturb bacterial redox homeostasis. While the molecular structure of Streptomyces coelicolor NsrR has been recently identified, the specific molecular structures of NsrR proteins in enterobacteria remain unknown. Our discovery of the crucial role of Val43 in the DNA recognition helix α3 of Salmonella NsrR offers valuable insights into the Hmp modulation under NO stress. Furthermore, the observed amino acid polymorphisms in the α3 helices of NsrR proteins across different bacterial species suggest the diverse evolution of NsrR structure and gene regulation in response to varying levels of NO pressure within their ecological niches.


Subject(s)
Nitric Oxide , Salmonella typhimurium , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Bacterial Proteins/metabolism , Bacteria/metabolism , Oxidation-Reduction , Gene Expression Regulation, Bacterial
2.
J Endocrinol ; 260(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38109258

ABSTRACT

We previously reported that Lactobacillus amylovorus KU4 (LKU4) promotes adipocyte browning in mice fed a high-fat diet (HFD mice) in part by remodeling the PPARγ transcription complex. However, the mechanism through which LKU4 enables PPARγ to drive adipocyte browning remains elusive. Here, we report that LKU4 inhibits the expression of PP4C in inguinal white adipose tissue of HFD mice and in insulin-resistant 3T3-L1 adipocytes, which promotes SIRT1-dependent PPARγ deacetylation by activating AMPK, leading to the browning of adipocytes. Consistently, the silencing of PP4C further enhances this pathway. Furthermore, we observed that lactate, a key LKU4 metabolite, reduces insulin-induced PP4C expression and suppresses PP4C inhibition of PPARγ deacetylation and transcriptional activity via AMPK-SIRT1, thereby facilitating the browning of adipocytes. Together, these data demonstrate that LKU4 promotes the AMPK-SIRT1-PPARγ pathway by inhibiting PP4C, thereby facilitating adipocyte browning in HFD mice.


Subject(s)
Insulins , Lactobacillus acidophilus , Mice , Animals , Mice, Obese , Sirtuin 1/genetics , Sirtuin 1/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , AMP-Activated Protein Kinases/metabolism , Obesity/metabolism , Adipose Tissue, White/metabolism , Insulins/metabolism , 3T3-L1 Cells , Diet, High-Fat
3.
Mol Nutr Food Res ; 67(20): e2200496, 2023 10.
Article in English | MEDLINE | ID: mdl-37650271

ABSTRACT

SCOPE: The present study aims to assess the protective effect of Lactobacillus johnsonii JNU3402 (LJ3402) against diet-induced non-alcoholic fatty liver disease (NAFLD) and determine the mechanism underlying its beneficial effect on the liver in mice. METHODS AND RESULTS: Seven-week-old male mice are fed a high-fat diet (HFD) with or without oral supplementation of LJ3402 for 14 weeks. In mice fed an HFD, LJ3402 administration alleviates liver steatosis, diet-induced obesity, and insulin resistance with a decreased hepatic expression of sterol-regulatory element-binding protein-1c (SREBP-1c), fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC), and an increased phosphorylation of SREBP-1c. The mechanistic study shows that LJ3402 inhibits SREBP-1c transcriptional activity by enhancing protein kinase A (PKA)-mediated phosphorylation and reduces the expression of its lipogenic target genes in AML12 and HepG2 cells, thereby attenuating hepatic lipid accumulation. Moreover, silencing the PKA α catalytic subunit or the inhibition of PKA activity by H89 abolishes LJ3402 suppression of free fatty acid (FFA)-induced SREBP-1c activity in hepatocytes. In addition, LJ3402 administration elevates the plasma lactate levels in mice fed an HFD; this lactate increases PKA-mediated SREBP-1c phosphorylation in AML12 cells with a decreased expression of its target genes, reducing hepatic lipid accumulation. CONCLUSION: LJ3402 attenuates HFD-induced fatty liver in mice through the lactate-PKA-SREBP-1c pathway.


Subject(s)
Lactobacillus johnsonii , Non-alcoholic Fatty Liver Disease , Male , Mice , Animals , Sterol Regulatory Element Binding Protein 1/metabolism , Lactobacillus johnsonii/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/metabolism , Lipid Metabolism , Diet, High-Fat/adverse effects , Fatty Acids, Nonesterified/pharmacology , Lactates , Mice, Inbred C57BL
5.
Cell Biosci ; 10: 119, 2020.
Article in English | MEDLINE | ID: mdl-33072258

ABSTRACT

BACKGROUND: TR4, an orphan nuclear receptor plays a key role in glucose and lipid metabolism by regulating the expression of genes involved in energy metabolism. We previously reported that overexpression of TR4 in 3T3-L1 adipocytes promotes lipid accumulation in part by facilitating fatty acid uptake and synthesis, indicating that TR4 tightly regulates lipid homeostasis during adipogenesis. Here, we report that ß-catenin suppresses TR4 transcriptional activity and that this inhibition is achieved through induction of Slug gene, a well-known transcription repressor in a variety of cells. METHODS: To generate the stable cell line, 3T3-L1 cells were transfected with plasmids then cultured in presence of geneticin and/or blasticidin for 2 weeks. The lipid accumulation was measured by Oil Red O. The TR4-Slug and TR4-ß-catenin interactions were checked by GST pull-down and mammalian two-hybrid assay. The TR4 transcriptional activities on various promoters were measured by luciferase activity. To check the binding affinity of TR4, we performed the gel shift and chromatin immunoprecipitation (ChIP) assay. Gene expression was detected by RT-qPCR at the mRNA level and western blotting at the protein level. RESULTS: Stable overexpression of Slug gene in 3T3-L1 preadipocytes strongly inhibited differentiation of 3T3-L1 preadipocytes. Using GST pull-down, gel shift and ChIP assays, we found that Slug abolished the formation of TR4 homodimers through direct interaction with TR4 and reduced the binding affinity of TR4 for its response elements located in TR4 target gene promoters such as fatty acid transport protein 1 and pyruvate carboxylase. Consistently, Slug inhibited TR4 target gene expression and was accompanied by repression of TR4-induced lipid accumulation in 3T3-L1 adipocytes. CONCLUSIONS: Our results demonstrated that Slug inhibits 3T3-L1 adipogenesis through suppression of TR4 transcriptional activity.

6.
Foods ; 9(10)2020 Oct 19.
Article in English | MEDLINE | ID: mdl-33086627

ABSTRACT

In this study, the role of non-viable Lactobacillus johnsonii JNU3402 (NV-LJ3402) in diet-induced obesity was investigated in mice fed a high-fat diet (HFD). To determine whether NV-LJ3402 exhibits a protective effect against diet-induced obesity, 7-week-old male C57BL/6J mice were fed a normal diet, an HFD, or an HFD with NV-LJ3402 for 14 weeks. NV-LJ3402 administration was associated with a significant reduction in body weight gain and in liver, epididymal, and inguinal white adipose tissue (WAT) and brown adipose tissue weight in HFD-fed mice. Concomitantly, NV-LJ3402 administration to HFD-fed mice also decreased the triglyceride levels in the plasma and metabolic tissues and slightly improved insulin resistance. Furthermore, NV-LJ3402 enhanced gene programming for energy dissipation in the WATs of HFD-fed mice as well as in 3T3-L1 adipocytes with increased peroxisome proliferator-activated receptor-γ (PPARγ) transcriptional activity, suggesting that the PPARγ pathway plays a key role in mediating the anti-obesity effect of NV-LJ3402 in HFD-fed mice. Furthermore, NV-LJ3402 administration in HFD-fed mice enhanced mitochondrial levels and function in WATs and also increased the body temperature upon cold exposure. Together, these results suggest that NV-LJ3402 could be safely used to develop dairy products that ameliorate diet-induced obesity and hyperlipidemia.

7.
Sci Rep ; 9(1): 20152, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882939

ABSTRACT

Browning of white adipose tissue (WAT) is currently considered a potential therapeutic strategy to treat diet-induced obesity. While some probiotics have protective effects against diet-induced obesity, the role of probiotics in adipose browning has not been explored. Here, we show that administration of the probiotic bacterium Lactobacillus amylovorus KU4 (LKU4) to mice fed a high-fat diet (HFD) enhanced mitochondrial levels and function, as well as the thermogenic gene program (increased Ucp1, PPARγ, and PGC-1α expression and decreased RIP140 expression), in subcutaneous inguinal WAT and also increased body temperature. Furthermore, LKU4 administration increased the interaction between PPARγ and PGC-1α through release of RIP140 to stimulate Ucp1 expression, thereby promoting browning of white adipocytes. In addition, lactate, the levels of which are elevated in plasma of HFD-fed mice following LKU4 administration, elicited the same effect on the interaction between PPARγ and PGC-1α in 3T3-L1 adipocytes, leading to a brown-like adipocyte phenotype that included enhanced Ucp1 expression, mitochondrial levels and function, and oxygen consumption rate. Together, these data reveal that LKU4 facilitates browning of white adipocytes through the PPARγ-PGC-1α transcriptional complex, at least in part by increasing lactate levels, leading to inhibition of diet-induced obesity.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat , Lactobacillus acidophilus/physiology , Obesity/etiology , Obesity/metabolism , PPAR gamma/metabolism , Probiotics , Signal Transduction , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Animals , Biomarkers , Cell Line , Diet, High-Fat/adverse effects , Disease Susceptibility , Gene Expression , Humans , Lipid Metabolism , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phenotype , Probiotics/administration & dosage , Protein Binding
8.
J Endocrinol ; 237(2): 87-100, 2018 05.
Article in English | MEDLINE | ID: mdl-29507043

ABSTRACT

Obesity is a major threat to public health, and it is strongly associated with insulin resistance and fatty liver disease. Here, we demonstrated that administration of Lactobacillus acidophilus NS1 (LNS1) significantly reduced obesity and hepatic lipid accumulation, with a concomitant improvement in insulin sensitivity, in high-fat diet (HFD)-fed mice. Furthermore, administration of LNS1 inhibited the effect of HFD feeding on the SREBP-1c and PPARα signaling pathways and reduced lipogenesis with an increase in fatty acid oxidation in ex vivo livers from HFD-fed mice. These LNS1 effects were confirmed in HepG2 cells and ex vivo livers by treatment with LNS1 culture supernatant (LNS1-CS). Interestingly, AMPK phosphorylation and activity in the liver of HFD-fed mice were increased by administration of LNS1. Consistently, chemical inhibition of AMPK with compound C, a specific inhibitor of AMPK, dramatically reduced the effect of LNS1-CS on lipid metabolism in HepG2 cells and ex vivo livers by modulating the SREBP-1c and PPARα signaling pathways. Furthermore, administration of LNS1 to HFD-fed mice significantly improved insulin resistance and increased Akt phosphorylation in the liver, white adipose tissue and skeletal muscle. Together, these data suggest that LNS1 may prevent diet-induced obesity and related metabolic disorders by improving lipid metabolism and insulin sensitivity through an AMPK→SREBP-1c/PPARα signaling pathway.


Subject(s)
Lactobacillus acidophilus/physiology , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/prevention & control , AMP-Activated Protein Kinases/metabolism , Animals , Diet, High-Fat/adverse effects , Hep G2 Cells , Humans , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Obesity/diet therapy , Obesity/etiology , Signal Transduction
9.
Korean J Food Sci Anim Resour ; 37(4): 529-534, 2017.
Article in English | MEDLINE | ID: mdl-28943765

ABSTRACT

Probiotics have been known to reduce high-fat diet (HFD)-induced metabolic diseases, such as obesity, insulin resistance, and type 2 diabetes. We recently observed that Lactobacillus acidophilus NS1 (LNS1), distinctly suppresses increase of blood glucose levels and insulin resistance in HFD-fed mice. In the present study, we demonstrated that oral administration of LNS1 with HFD feeding to mice significantly reduces hepatic expression of phosphoenolpyruvate carboxykinase (PEPCK), a key enzyme in gluconeogenesis which is highly increased by HFD feeding. This suppressive effect of LNS1 on hepatic expression of PEPCK was further confirmed in HepG2 cells by treatment of LNS1 conditioned media (LNS1-CM). LNS1-CM strongly and specifically inhibited HNF4α-induced PEPCK promoter activity in HepG2 cells without change of HNF4α mRNA levels. Together, these data demonstrate that LNS1 suppresses PEPCK expression in the liver by regulating HNF4α transcriptional activity, implicating its role as a preventive or therapeutic approach for metabolic diseases.

10.
Mol Cell Endocrinol ; 423: 22-9, 2016 Mar 05.
Article in English | MEDLINE | ID: mdl-26762765

ABSTRACT

In this study, we show that reduction of glucose concentration increases TR4 expression in 3T3-L1 cells via stimulation of the GSK-3ß-CREB pathway. While GSK-3ß and CREB increased TR4 expression in 3T3-L1 cells, inhibition of CREB expression or activity resulted in loss of GSK-3ß-mediated enhancement of TR4 expression. In addition, CREB enhanced murine TR4 promoter activity via direct binding to a cAMP response element located in the promoter, and this CREB effect was further strengthened by GSK-3ß. Moreover, silencing of TR4 expression by a gene-specific microRNA inhibited CREB-induced lipid accumulation in 3T3-L1 adipocytes, suggesting that TR4 could be a key mediator of CREB-induced lipogenesis.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Glycogen Synthase Kinase 3/physiology , Toll-Like Receptor 4/metabolism , 3T3-L1 Cells , Animals , Energy Metabolism , Gene Expression Regulation , Glucose/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Homeostasis , Humans , Mice , Protein Binding , Response Elements , Signal Transduction , Toll-Like Receptor 4/genetics , Transcriptional Activation
11.
FEBS Lett ; 588(21): 3947-53, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25240193

ABSTRACT

We show that testicular orphan nuclear receptor 4 (TR4) increases the expression of pyruvate carboxylase (PC) gene in 3T3-L1 adipocytes by direct binding to a TR4 responsive element in the murine PC promoter. While TR4 overexpression increased PC activity, oxaloacetate (OAA) and glycerol levels with enhanced incorporation of (14)C from (14)C-pyruvate into fatty acids in 3T3-L1 adipocytes, PC knockdown by short interfering RNA (siRNA) or inhibition of PC activity by phenylacetic acid (PAA) abolished TR4-enhanced fatty acid synthesis. Moreover, TR4 microRNA reduced PC expression with decreased fatty acid synthesis in 3T3-L1 adipocytes, suggesting that TR4-mediated enhancement of fatty acid synthesis in adipocytes requires increased expression of PC gene.


Subject(s)
Adipocytes/metabolism , Fatty Acids/biosynthesis , Gene Expression Regulation, Enzymologic , Pyruvate Carboxylase/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , 3T3-L1 Cells , Animals , Gene Knockdown Techniques , Lipogenesis , Mice , Promoter Regions, Genetic/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/genetics
12.
Endocr Relat Cancer ; 21(3): R279-300, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24623743

ABSTRACT

Peroxisome proliferator-activated receptor γ (PPARγ, NR1C3) and testicular receptor 4 nuclear receptor (TR4, NR2C2) are two members of the nuclear receptor (NR) superfamily that can be activated by several similar ligands/activators including polyunsaturated fatty acid metabolites, such as 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, as well as some anti-diabetic drugs such as thiazolidinediones (TZDs). However, the consequences of the transactivation of these ligands/activators via these two NRs are different, with at least three distinct phenotypes. First, activation of PPARγ increases insulin sensitivity yet activation of TR4 decreases insulin sensitivity. Second, PPARγ attenuates atherosclerosis but TR4 might increase the risk of atherosclerosis. Third, PPARγ suppresses prostate cancer (PCa) development and TR4 suppresses prostate carcinogenesis yet promotes PCa metastasis. Importantly, the deregulation of either PPARγ or TR4 in PCa alone might then alter the other receptor's influences on PCa progression. Knocking out PPARγ altered the ability of TR4 to promote prostate carcinogenesis and knocking down TR4 also resulted in TZD treatment promoting PCa development, indicating that both PPARγ and TR4 might coordinate with each other to regulate PCa initiation, and the loss of either one of them might switch the other one from a tumor suppressor to a tumor promoter. These results indicate that further and detailed studies of both receptors at the same time in the same cells/organs may help us to better dissect their distinct physiological roles and develop better drug(s) with fewer side effects to battle PPARγ- and TR4-related diseases including tumor and cardiovascular diseases as well as metabolic disorders.


Subject(s)
Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , PPAR gamma/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/prevention & control , Animals , Gene Expression Regulation , Humans , Male
13.
J Agric Food Chem ; 61(19): 4577-84, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23651430

ABSTRACT

Catechin polymers were produced by laccase (12 U/mL) in a mixture of sodium acetate buffer (1% (+)-catechin, 100 mM, pH 5) and methanol (buffer:methanol = 95:5, v/v). The freeze-dried catechin polymers were recovered from the precipitate after dialysis followed by centrifugation. Catechin polymers extracted with 20% ethanol had potent inhibitory activity against α-glucosidase with an IC50 value of 4 µg/mL, and they were present as a mixture of dimers, trimers, and tetramers. The antihyperglycemic effect of the catechin polymers was confirmed by an oral maltose tolerance test. The catechin polymers also had significantly improved antiglycation and superoxide dismutase-like activities compared to those of (+)-catechin. Since formation of advanced glycation end products and oxidative stress are accelerated in hyperglycemic conditions, we suggest that enzymatic production of catechin polymers could have a potential protective effect in type 2 diabetes, diabetic complications, and other free radical related diseases.


Subject(s)
Catechin/pharmacology , Glycation End Products, Advanced/metabolism , Glycoside Hydrolase Inhibitors , Laccase/metabolism , Animals , Biflavonoids/chemistry , Catalysis , Catechin/chemical synthesis , Catechin/chemistry , Chromatography, Liquid , Freeze Drying , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Inhibitory Concentration 50 , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Molecular Weight , Oxidative Stress/drug effects , Polymers/chemical synthesis , Polymers/pharmacology , Superoxide Dismutase/metabolism , alpha-Glucosidases/metabolism
14.
Biomaterials ; 32(35): 9401-14, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21911251

ABSTRACT

Biomedical applications of magnetic nanoparticles (MNP), including superparamagnetic nanoparticles, have expanded dramatically in recent years. Systematic and standardized cytotoxicity assessment to ensure the biosafety and biocompatibility of those applications is compulsory. We investigated whether exposure to static magnetic field (SMF) from e.g. magnetic resonance imaging (MRI) could affect the cytotoxicity of superparamagnetic iron oxide (SPIO) nanoparticles using mouse hepatocytes and ferucarbotran, a liver-selective MRI contrast agent as a model system. We show that while the SPIO satisfied the conventional cytotoxicity assessment, clinical doses combined with SMF exposure exerts synergistic adverse effects such as reduced cell viability, apoptosis, and cell cycle aberrations on hepatocytes in vitro and in vivo. Concomitant treatments with the SPIO and SMF generated SPIO aggregates, which demonstrated enhanced cellular uptake, was sufficient to induce the cytotoxicity without further SMF, emphasizing that the SPIO aggregates were the predominant source of the cytotoxicity. Interestingly, the apoptotic effect was dependent on levels of reactive oxygen species (ROS) and SPIO uptake while the reduced cell viability was independent of these factors. Moreover, long-term monitoring showed a significant increase in multinuclear giant cells in the cells concomitantly treated with the SPIO and SMF compared with the control. The results demonstrate that the SPIO produces unidentified cytotoxicity on liver in the presence of SMF and the SPIO aggregates predominantly exert the effect. Since aggregation of MNP in biological milieu in the presence of strong SMF is inevitable, a fundamentally different approach to surface fabrication is essential to increase the biocompatibility of MNP.


Subject(s)
Magnetic Fields , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity , Animals , Cell Death/drug effects , Cell Line , Dextrans/chemistry , Dextrans/toxicity , Endocytosis/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Magnetic Resonance Imaging , Magnetite Nanoparticles/ultrastructure , Mice , Reactive Oxygen Species/metabolism
15.
FEBS Lett ; 585(17): 2763-7, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21843524

ABSTRACT

We show that TR4 facilitates lipid accumulation in 3T3-L1 adipocytes via induction of the FATP1 gene. Further study showed that TR4 transactivated FATP1 5' promoter activity via direct binding to the TR4 responsive element located at the FATP1 5' promoter region. Constitutive overexpression of TR4 in 3T3-L1 adipocytes resulted in increased lipid accumulation, accompanied by an increase in fatty acid uptake. However, small interfering RNA knockdown of FATP1 abolished TR4-enhanced fatty acid uptake. Moreover, microRNA-mediated silencing of TR4 in 3T3-L1 adipocytes drastically reduced basal FATP1 5' promoter activity and FATP1 expression with reduced lipid accumulation.


Subject(s)
Adipocytes/metabolism , Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism , 3T3-L1 Cells , Animals , Blotting, Western , CD36 Antigens/genetics , CD36 Antigens/metabolism , Chromatin Immunoprecipitation , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Fatty Acid Transport Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Mice , MicroRNAs/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Diabetes ; 60(5): 1493-503, 2011 May.
Article in English | MEDLINE | ID: mdl-21478464

ABSTRACT

OBJECTIVE: TR4 is a nuclear receptor without clear pathophysiological roles. We investigated the roles of hepatic TR4 in the regulation of lipogenesis and insulin sensitivity in vivo and in vitro. RESEARCH DESIGN AND METHODS: TR4 activity and phosphorylation assays were carried out using hepatocytes and various TR4 wild-type and mutant constructs. Liver tissues from TR4 knockout, C57BL/6, and db/db mice were examined to investigate TR4 target gene stearoyl-CoA desaturase (SCD) 1 regulation. RESULTS: TR4 transactivation is inhibited via phosphorylation by metformin-induced AMP-activated protein kinase (AMPK) at the amino acid serine 351, which results in the suppression of SCD1 gene expression. Additional mechanistic dissection finds TR4-transactivated SCD1 promoter activity via direct binding to the TR4-responsive element located at -243 to -255 on the promoter region. The pathophysiological consequences of the metformin→AMPK→TR4→SCD1 pathway are examined via TR4 knockout mice and primary hepatocytes with either knockdown or overexpression of TR4. The results show that the suppression of SCD1 via loss of TR4 resulted in reduced fat mass and increased insulin sensitivity with increased ß-oxidation and decreased lipogenic gene expression. CONCLUSIONS: The pathway from metformin→AMPK→TR4→SCD1→insulin sensitivity suggests that TR4 may function as an important modulator to control lipid metabolism, which sheds light on the use of small molecules to modulate TR4 activity as a new alternative approach to battle the metabolic syndrome.


Subject(s)
Liver/metabolism , Metformin/pharmacology , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Stearoyl-CoA Desaturase/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Glucose Tolerance Test , Immunoprecipitation , Insulin/pharmacology , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Stearoyl-CoA Desaturase/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
17.
Mol Cells ; 31(4): 371-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21331774

ABSTRACT

Stearoyl-CoA desaturase (SCD), the rate-limiting enzyme in the biosynthesis of monounsaturated fatty acids, is highly expressed in prostate cancer although the SCD protein has been known to be rapidly turned over by proteolytic cleavage. The present data demonstrate that SCD can promote proliferation of androgen receptor (AR)-positive LNCaP prostate cancer cells and enhance dihydrotestosterone (DHT)-induced AR transcriptional activity, resulting in increased expression of prostate-specific antigen (PSA) and kallikrein-related peptidase 2 (KLK2). Interestingly, among the previously reported SCD-derived peptides produced by proteolytic cleavage of SCD, a peptide spanning amino acids 130-162 of SCD (SCD-CoRNR) contained the CoRNR box motif (LFLII) and enhanced AR transcriptional activity. In contrast, a mutant SCD-CoRNR in which Leu136 was replaced by Ala had no effect on AR transcriptional activity. Moreover, SCD-CoRNR directly interacted with AR and inhibited RIP140 suppression of AR transactivation. Knockdown of the SCD gene by SCD microRNA suppressed AR transactivation with decreased cell proliferation, suggesting that SCD may regulate the proliferation of LNCaP cells via modulation of AR transcriptional activity. Moreover, ectopic expression of SCD in LNCaP cells facilitated LNCaP tumor formation and growth in nude mice. Together, the data indicate that SCD plays a key role in the regulation of AR transcriptional activity in prostate cancer cells.


Subject(s)
Cell Proliferation , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Stearoyl-CoA Desaturase/metabolism , Transcriptional Activation , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Male , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Nuclear Proteins/antagonists & inhibitors , Nuclear Receptor Interacting Protein 1 , Prostatic Neoplasms/pathology , RNA Interference , Receptors, Androgen/metabolism , Sequence Alignment , Stearoyl-CoA Desaturase/genetics , Transcription, Genetic , Tumor Burden , Up-Regulation
18.
Proc Natl Acad Sci U S A ; 106(32): 13353-8, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19666541

ABSTRACT

Testicular orphan nuclear receptor 4 (TR4) is an orphan member of the nuclear receptor superfamily with diverse physiological functions. Using TR4 knockout (TR4(-/-)) mice to study its function in cardiovascular diseases, we found reduced cluster of differentiation (CD)36 expression with reduced foam cell formation in TR4(-/-) mice. Mechanistic dissection suggests that TR4 induces CD36 protein and mRNA expression via a transcriptional regulation. Interestingly, we found this TR4-mediated CD36 transactivation can be further enhanced by polyunsaturated fatty acids (PUFAs), such as omega-3 and -6 fatty acids, and their metabolites such as 15-hydroxyeico-satetraonic acid (15-HETE) and 13-hydroxy octa-deca dieonic acid (13-HODE) and thiazolidinedione (TZD)-rosiglitazone. Both electrophoretic mobility shift assays (EMSA) and chromatin immunoprecipitation (ChIP) assays demonstrate that TR4 binds to the TR4 response element located on the CD36 5'-promoter region for the induction of CD36 expression. Stably transfected TR4-siRNA or functional TR4 cDNA in the RAW264.7 macrophage cells resulted in either decreased or increased CD36 expression with decreased or increased foam cell formation. Restoring functional CD36 cDNA in the TR4 knockdown macrophage cells reversed the decreased foam cell formation. Together, these results reveal an important signaling pathway controlling CD36-mediated foam cell formation/cardiovascular diseases, and findings that TR4 transactivation can be activated via its ligands/activators, such as PUFA metabolites and TZD, may provide a platform to screen new drug(s) to battle the metabolism syndrome, diabetes, and cardiovascular diseases.


Subject(s)
CD36 Antigens/metabolism , Cell Nucleus/metabolism , Fatty Acids, Unsaturated/metabolism , Foam Cells/cytology , Foam Cells/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Base Sequence , CD36 Antigens/genetics , Cell Nucleus/drug effects , Foam Cells/drug effects , Ligands , Mice , Molecular Sequence Data , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Rosiglitazone , Thiazolidinediones/pharmacology , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
19.
Mol Cells ; 26(4): 409-14, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18562803

ABSTRACT

The cholesteryl ester transfer protein (CETP), a key player in cholesterol metabolism, has been shown to promote the transfer of triglycerides from very low density lipoprotein (VLDL) and low density lipoprotein (LDL) to high density lipoprotein (HDL) in exchange for cholesterol ester. Here we demonstrate that farnesoid X receptor alpha (FXRalpha; NR1H4) down-regulates CETP expression in HepG2 cells. A FXRalpha ligand, chenodeoxycholic acid (CDCA), suppressed basal mRNA levels of the CETP gene in HepG2 cells in a dose-dependent manner. Using gel shift and chromatin immunoprecipitation (ChIP) assays, we found that FXRalpha could bind to the liver X receptor alpha (LXRalpha; NR1H3) binding site (LXRE; DR4RE) located within the CETP 5' promoter region. FXRalpha suppressed LXRalpha-induced DR4RE-luciferase activity and this effect was mediated by a binding competition between FXRalpha and LXRalpha for DR4RE. Furthermore, the addition of CDCA together with a LXRalpha ligand, GW3965, to HepG2 cells was shown to substantially decrease mRNA levels of hepatic CETP gene, which is typically induced by GW3965. Together, our data demonstrate that FXRalpha down-regulates CETP gene expression via binding to the DR4RE sequence within the CETP 5' promoter and this FXRalpha binding is essential for FXRalpha inhibition of LXRalpha-induced CETP expression.


Subject(s)
Cholesterol Ester Transfer Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transcription Factors/metabolism , Binding, Competitive/drug effects , Cell Line , Chenodeoxycholic Acid/pharmacology , Down-Regulation/drug effects , Humans , Liver X Receptors , Orphan Nuclear Receptors , Protein Binding/drug effects , Protein Multimerization/drug effects , Response Elements , Retinoid X Receptor alpha/metabolism , Signal Transduction/drug effects
20.
Diabetes ; 56(12): 2901-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17827404

ABSTRACT

OBJECTIVE: Regulation of phosphoenolpyruvate carboxykinase (PEPCK), the key gene in gluconeogenesis, is critical for glucose homeostasis in response to quick nutritional depletion and/or hormonal alteration. RESEARCH DESIGN/METHODS AND RESULTS: Here, we identified the testicular orphan nuclear receptor 4 (TR4) as a key PEPCK regulator modulating PEPCK gene via a transcriptional mechanism. TR4 transactivates the 490-bp PEPCK promoter-containing luciferase reporter gene activity by direct binding to the TR4 responsive element (TR4RE) located at -451 to -439 in the promoter region. Binding to TR4RE was confirmed by electrophoretic mobility shift and chromatin immunoprecipitation assays. Eliminating TR4 via knockout and RNA interference (RNAi) in hepatocytes significantly reduced the PEPCK gene expression and glucose production in response to glucose depletion. In contrast, ectopic expression of TR4 increased PEPCK gene expression and hepatic glucose production in human and mouse hepatoma cells. Mice lacking TR4 also display reduction of PEPCK expression with impaired gluconeogenesis. CONCLUSIONS: Together, both in vitro and in vivo data demonstrate the identification of a new pathway, TR4 --> PEPCK --> gluconeogenesis --> blood glucose, which may allow us to modulate metabolic programs via the control of a new key player, TR4, a member of the nuclear receptor superfamily.


Subject(s)
Gluconeogenesis/physiology , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Animals , Cells, Cultured , Gene Expression Regulation , Hepatocytes/physiology , Male , Plasmids , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA Interference , Rats , Receptors, Steroid/deficiency , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/physiology , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...