Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biol Pharm Bull ; 42(10): 1628-1636, 2019.
Article in English | MEDLINE | ID: mdl-31582651

ABSTRACT

Cigarette smoke extract (CSE) contains many toxicants and may derange the physiological processes, such as cholesterol metabolism. We examined the impact of CSE on transcriptional regulation mediated peroxisome proliferator-activated receptors (PPARs) and its interaction with cofactors to elucidate differences in the molecular mechanism between CSE and other agonists of PPARs. We constructed several mutant PPARs (mPPARs) with amino acid substitution in the ligand-binding domain, which according to the molecular modeling, may affect the binding of agonists. In transient expression assays, each wild-type peroxisome proliferator-activated receptor (PPAR) mediated transcription stimulated by CSE was faintly yet significantly elevated compared to the control. The CSE-induced transcriptional activation was abolished in the H323A, H323Y, S342A, and H449A mPPARγs, although the activation elevated by pioglitazone was reserved. In the mPPARγ with Y473A and mPPARß/δs with H286Y and Y436A, the pioglitazone-induced or L165041-activated transcriptional elevations were decreased and were lower than that of CSE-induced stimulation. These results suggested that CSE activated both mutant PPARs to be selectively different from those ligands. Mammalian two-hybrid assay illustrated that CSE could mildly recruit SRC1 or GRIP1 to the wild-type PPARγ. Representative ingredients, such as acrolein and crotonaldehyde present in CSE, could stimulate PPAR isoforms even at the toxicological concentrations and might possibly contribute to stimulatory effects. CSE mildly regulates the cholesterol metabolism-related genes, such as low density lipoprotein (LDL) receptor and Liver X receptor (LXR)ß. In conclusion, these CSE effects the nuclear hormone receptors and their cofactors thereby disturbing metabolic phenomena. Therefore, CSE might be involved in cholesterol metabolism.


Subject(s)
Nicotiana , Peroxisome Proliferator-Activated Receptors/metabolism , Smoke , Amino Acid Substitution , Cell Line , Cholesterol, LDL/metabolism , Humans , Liver X Receptors/genetics , Peroxisome Proliferator-Activated Receptors/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, LDL/genetics
2.
Growth Horm IGF Res ; 46-47: 24-35, 2019.
Article in English | MEDLINE | ID: mdl-31158782

ABSTRACT

OBJECTIVE: The objective of our study was to examine the direct action of insulin-like growth factor-1(IGF-1) signaling on energy homeostasis in myocytes. DESIGN: We studied the IGF-1 stimulation of mitochondrial uncoupling protein 3 (UCP3) expression in the HEK 293 derived cell line TSA201, murine C2C12 skeletal muscle myoblasts, and rat L6 skeletal myoblasts. We also investigated the direct effect of IGF-1 on the Insulin/IGF-1 receptor (IGF-1R)/phosphatidylinositol 3 (PI3)-Akt/forkhead box O4 (FOXO4) pathway using a combination of a reporter assay, semi-quantitative polymerase chain reaction, western blotting, and animal experiments. RESULTS: We demonstrated that IGF-1 regulates UCP3 expression via phosphorylation of FOXO4, which is a downstream signal transducer of IGF-1. UCP3 expression increased with activated FOXO4 in a dose-dependent manner. We also examined the functional FOXO4 binding site consensus sequences and identified it as the -1922 bp site in the UCP3 promoter region. UCP3 was also found to be concomitantly expressed with IGF-1 during differentiation of C2C12 myoblasts. Our animal experiments showed that high fat diet induced IGF-1 levels which likely influenced UCP3 expression in the skeletal muscle. CONCLUSION: Our findings demonstrate that that IGF-1 directly stimulates UCP3 expression via the IGF-1/IGF-1R/PI3-Akt/FOXO4 pathway.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Insulin-Like Growth Factor I/pharmacology , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Uncoupling Protein 3/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , Forkhead Transcription Factors/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Uncoupling Protein 3/genetics
3.
Growth Factors ; 37(5-6): 247-256, 2019 12.
Article in English | MEDLINE | ID: mdl-32156173

ABSTRACT

Mitochondria uncoupling protein2 (UCP2) expressed ubiquitously is a key molecule of energy metabolism. Insulin-like growth factor-1 (IGF-1) is a hormone, a target molecule of growth hormone (GH) signal pathway, which is also known as the drug "mecasermin" for clinical usages. IGF-1 is seemed to be closely related to metabolic diseases, such as adult GH deficiency. However, there has not been reports depicted possible relationship with each other. So, we sought to elucidate the mechanisms by which expression of UCP2 is regulated by IGF-1 via FOXO1. The findings suggested that three sequences in the consensus UCP2 promoter play complementary functional roles in the functional expression of FOXO1. So, we found that FOXO1 is involved in IGF-1-mediated energy metabolism greater than that of direct action of GH via STAT5. Our findings suggested that IGF-1 was involved in energy metabolism by regulating the expression of UCP2 via the PI3K/Akt/FOXO1 pathway.


Subject(s)
Forkhead Box Protein O1/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Uncoupling Protein 2/metabolism , 3T3-L1 Cells , Adipose Tissue/metabolism , Animals , Energy Metabolism , Gene Expression Regulation , HEK293 Cells , Hep G2 Cells , Humans , Mice , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Promoter Regions, Genetic , Receptor, IGF Type 1/metabolism , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism
4.
Biol Pharm Bull ; 41(3): 383-393, 2018.
Article in English | MEDLINE | ID: mdl-29491215

ABSTRACT

Cigarette smoke contains over 4800 compounds, including at least 200 toxicants or endocrine disruptors. Currently, effects of cigarette smoke on thyroid hormone (TH) levels remains to be clarified. Here, we demonstrate that cigarette smoke extract (CSE) possesses thyroid hormone properties and acts synergistically as a partial agonist for thyroid hormone receptors (TRs) in the presence of TH. In transient gene expression experiments, CSE stimulated transcriptional activity with TH in a dose-dependent manner. Stimulatory effects were observed with physiological TH concentrations, although CSE did not activate TRs without TH. CSE (5%) dissolved in phosphate-buffered saline (PBS) supplemented with 1 nM TH was approximately comparable to 3.2±0.1 and 2.3±0.2 nM of TRα1 and TRß1, respectively. To illustrate probable mechanisms of the CSE agonistic activity, effects on TR mediated transcriptional functions with cofactors were investigated. With a mammalian two-hybrid assay, CSE recruited the nuclear coactivators glucocorticoid receptor interacting protein 1 (GRIP1) and steroid receptor coactivator 1 (SRC1) to the TR. Unsaturated carbonyl compounds, acrolein, crotonaldehyde, and methyl vinyl ketone, representative constituents of CSE, retained such agonistic properties and possibly contributed to stimulatory effects. The results suggest that CSE recruits a transcriptional activator and may reinforce TH binding to the TR additively, resulting in gene expression. CSE partially agonizes TH action and may disturb the function of various nuclear hormone receptor types and their cofactors to disrupt the physiological processes.


Subject(s)
Nicotiana/adverse effects , Receptors, Thyroid Hormone/drug effects , Smoke/adverse effects , Thyroid Hormones/pharmacology , Transcription, Genetic/drug effects , Carrier Proteins/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Malate Dehydrogenase/biosynthesis , Nerve Tissue Proteins/drug effects , Nuclear Receptor Coactivator 1/genetics , Receptors, Thyroid Hormone/genetics , Smoke/analysis , Thyroid Hormone Receptors alpha/drug effects , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/drug effects , Thyroid Hormone Receptors beta/genetics , Nicotiana/chemistry
5.
Growth Horm IGF Res ; 40: 44-54, 2018 06.
Article in English | MEDLINE | ID: mdl-29398371

ABSTRACT

OBJECTIVE: We evaluated the direct action of GH signaling in energy homeostasis in myocytes. DESIGN: We investigated the GH-induced expression of UCP3 in human embryonic kidney 293 cells, human H-EMC-SS chondrosarcoma cells, murine C2C12 skeletal muscle myoblasts, and rat L6 skeletal muscle cells, as well as its direct effect on the GHR/JAK/STAT5 pathway using a combination of a reporter assay, real-time quantitative polymerase chain reaction, and western blotting. RESULTS: We demonstrated that the regulation of energy metabolism by GH involves UCP3 via activated STAT5, a signal transducer downstream of GH. UCP3 expression increased with STAT5 in a dose-dependent manner and was higher than that of UCP2. We confirmed the functional STAT5 binding site consensus sequences at -861 and -507 bp in the UCP3 promoter region. CONCLUSION: The results suggest that GH stimulates UCP3 directly and that UCP2 and that UCP3 participate in the signal transduction pathway that functions downstream of the GHR/JAK/STAT.


Subject(s)
Chondrosarcoma/metabolism , Gene Expression Regulation/drug effects , Growth Hormone/pharmacology , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Uncoupling Protein 3/metabolism , Animals , Cells, Cultured , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Muscle, Skeletal/cytology , Myoblasts, Skeletal/cytology , Rats , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction , Uncoupling Protein 3/genetics
6.
Growth Horm IGF Res ; 37: 31-39, 2017 12.
Article in English | MEDLINE | ID: mdl-29111497

ABSTRACT

OBJECTIVE: The transition of white adipocytes to beige cells (a phenomenon referred to as browning or beigeing) during obesity has been previously reported. Our study aimed to examine the mechanisms through which obesity induced by a high fat diet (HFD) affects uncoupling protein 1 (UCP1) expression via signal transduction and activator of transcription 5 (STAT5s). DESIGN: Seven-week-old male C57BL/6J mice were fed a normal or HFD for 11weeks. Body weight, white adipose tissue weight, and blood lipid and glucose levels were measured. To unveil the molecular mechanisms of UCP1 expression in adipose tissue, we performed further studying 3T3-L1 cells using qRT-PCR. We also measured UCP1 promoter activity in the TSA201 cell line using a dual luciferase assay. In addition, we analyzed the predicted consensus sequences for STAT5 binding in the UCP1 promoter region. RESULTS: Mice fed an HFD had higher body weight and intra-abdominal adipose tissues weight and a higher expression of UCP1, GH receptor (GHR), STATs, suppressors of cytokine signaling (SOCSs), and cytokine-inducible SH2-containing protein (CISH) compared to control mice. In 3T3-L1 cell studies, GH induced phosphorylation of the STAT5, SOCSs, CISH and UCP1 expressions. UCP1 promoter activity was associated with constitutively active STAT5 in a dose-dependent manner. We confirmed functional STAT5 binding sites at -425, -279, and -178bp of the UCP1 promoter. CONCLUSION: We suggest that endogenous GH induces UCP1 expression in adipose tissue via STAT5.


Subject(s)
Adipose Tissue, White/metabolism , Growth Hormone/metabolism , STAT5 Transcription Factor/genetics , Uncoupling Protein 1/metabolism , 3T3-L1 Cells , Abdominal Fat/metabolism , Adipocytes/metabolism , Animals , Binding Sites , Blood Glucose/metabolism , Body Weight , Calcium Phosphates/metabolism , Diet, High-Fat , HEK293 Cells , Humans , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Phosphorylation , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Messenger/metabolism , Uncoupling Protein 1/genetics
7.
J Biomed Sci ; 23(1): 56, 2016 Jul 29.
Article in English | MEDLINE | ID: mdl-27473111

ABSTRACT

BACKGROUND: The impact of vitamin D3 (VD3) on obesity has been reported in the past. Our study was aimed at investigating the possible mechanisms by which VD3 affects obesity induced by a high fat diet. METHODS: Eight-week-old C57BL/6 J male mice were fed a normal- or high-fat diet for 9 weeks and were treated with a gavage of vehicle (corn oil) or cholecalciferol (50 µg/kg, daily). Body weight, white adipose tissue weight, blood lipid and glucose levels were measured. In addition, we investigated the expression of 1,25(OH)2D3 (calcitriol)/VDR-regulated genes involved in energy and lipid metabolism, such as of uncoupling protein 3 (UCP3), by using qRT-PCR in the liver, adipose tissue, skeletal muscle and C2C12, L6, and H-EMC-SS cells. We also measured UCP3 promoter transcription in the same cell lines using a Dual Luciferase Assay. Furthermore, we analyzed the binding site consensus sequences of VDR on the UCP3 promoter. RESULTS: Mice consuming a high-fat diet treated with cholecalciferol had lower body weight and adipose tissue weight and higher expression of UCP3 compared to the other treatment groups. Changes in the expression of genes correlated with calcitriol/VDR. Luciferase activity was dose-dependently associated with calcitriol/VDR levels. We confirmed the functional VDR binding site consensus sequences at -2200, -1561, -634, and +314 bp in the UCP3 promoter region. CONCLUSION: We suggest that VD3/VDR inhibits weight gain by activating UCP3 in the muscles.


Subject(s)
Calcitriol/pharmacology , Cholecalciferol/pharmacology , Gene Expression Regulation/drug effects , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Obesity/metabolism , Receptors, Calcitriol/metabolism , Uncoupling Protein 3/biosynthesis , Animals , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Male , Mice , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Receptors, Calcitriol/genetics , Uncoupling Protein 3/genetics
8.
J Mol Endocrinol ; 57(1): 23-32, 2016 07.
Article in English | MEDLINE | ID: mdl-27154546

ABSTRACT

The active form of vitamin D3 (1α,25(OH)2D3, also known as calcitriol) controls the expression of target genes via the vitamin D receptor (VDR). Vitamin D-dependent rickets type II (VDDRII) is a congenital disease caused by inactivating mutations in the VDR The condition is treated with high doses of calcitriol, but the therapeutic effects of other synthetic VD3 analogs have not yet been investigated. In the present study, we analyzed the transcriptional activity of seven different VD3 analogs with VDRs carrying ligand-binding domain mutations identified in VDDRII patients. Wild-type VDR (WT-VDR) and seven mutant VDRs were expressed in TSA201 human embryonic kidney cells, HepG2 human liver cancer cells, and MC3T3-E1 mouse calvaria cells, and their transcriptional activation with VD3 analogs were analyzed by performing transient expression assays, western blotting, and quantitative real-time PCR. The results demonstrated that falecalcitriol stimulated significantly higher transcriptional activation of the WT-VDR and some mutant VDRs than did calcitriol. Calcitriol showed almost no transcriptional activation of the VDR with the I268T mutation identified in a severe case of VDDRII, whereas falecalcitriol caused a dose-dependent increase in the activation of this mutant VDR. Our findings demonstrate that falecalcitriol has a VDR activation profile distinct from that of calcitriol and may exhibit therapeutic effects even on difficult-to-treat VDDRII cases resistant to calcitriol. It is also possible that VDDRII patients responding to high doses of calcitriol could be appropriately treated with low doses of falecalcitriol.


Subject(s)
Cholecalciferol/pharmacology , Gene Expression Regulation/drug effects , Mutation , Receptors, Calcitriol/genetics , Transcription, Genetic , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Animals , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cell Line , Cholecalciferol/analogs & derivatives , Humans , Mice , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcitriol/metabolism
9.
Growth Horm IGF Res ; 29: 57-62, 2016 08.
Article in English | MEDLINE | ID: mdl-27150070

ABSTRACT

OBJECTIVE: To determine if and how growth hormone (GH) signaling is involved in energy metabolism. DESIGN: We used human embryonic kidney TSA201 cells, human H-EMC-SS chondrosarcoma cells, rat L6 skeletal muscle cells, and murine C2C12 skeletal muscle myoblasts to investigate GH-induced expression of uncoupling protein2 (UCP2) to the GHR/JAK/STAT5 pathway by a combination of a reporter assay, electrophoretic mobility shift assay (EMSA), real-time quantitative PCR, Western blotting. RESULTS: We demonstrated that the regulation energy metabolism, which was hypothesized to be directly acted on by GH, involves UCP2 via activated STAT5B, a signal transducer downstream of GH. We also showed that the sequence at the -586 'TTCnGA' may function as a novel putative consensus sequence of STAT5s. CONCLUSION: The results suggest that GH regulates energy metabolism directly in myocytes and that UCP2 participates in the signal transduction pathway that functions downstream of the GHR/JAK/STAT.


Subject(s)
Chondrosarcoma/metabolism , Energy Metabolism/genetics , Growth Hormone/metabolism , Janus Kinase 2/metabolism , Muscle Fibers, Skeletal/metabolism , STAT5 Transcription Factor/metabolism , Uncoupling Protein 2/genetics , Animals , Blotting, Western , Cell Line, Tumor , Chondrosarcoma/genetics , Electrophoretic Mobility Shift Assay , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Janus Kinases/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptors, Somatotropin/metabolism , Signal Transduction
10.
Endocr Res ; 41(1): 34-42, 2016.
Article in English | MEDLINE | ID: mdl-26513165

ABSTRACT

Thyroid hormone exerts a pleiotropic effect on development, differentiation, and metabolism through thyroid hormone receptor (TR). A novel thyroid hormone receptor ß isoform (TRß4) was cloned using PCR from a human pituitary cDNA library as a template. We report here the characterization of TRß4 from a molecular basis. Temporal expression of TRß4 during the fetal period is abundant in the brain and kidney, comparable with the adult pattern. Western blot analysis revealed that TRs are ubiquitination labile proteins, while TRß1 is potentially stable. TRß1, peroxisome proliferator-activated receptors (PPAR), and vitamin D receptor (VDR), which belong to class II transcription factors that function via the formation of heterodimeric complexes with retinoid X receptor (RXR), were suppressed by TRß4 in a dose-dependent manner. Thus, TRß4 exhibits ligand-independent transcriptional silencing, possibly as a substitute for dimerized RXR. In this study, TRß1 and TRß4 transcripts were detected in several cell lines. Quantitative RT-PCR assay showed that the expression of TRß4 in human embryonic carcinoma cells of the testis was suppressed by sex hormone in a reciprocal manner to TRß1. In contrast, TRß4 was expressed under a high dose of triiodothyronine (T3) in a reciprocal manner to TRß1. Finally, in transiently transfected NIH-3T3 cells, green fluorescence protein (GFP)-tagged TRß4 was mostly nuclear in both the absence and the presence of T3. By mutating defined regions of both TRßs, we found that both TRß1 and TRß4 had altered nuclear/cytoplasmic distribution as compared with wild-type, and different to T3 and the nuclear receptor corepressor (NCoR). Thus, site-specific DNA binding is not essential for maintaining TRßs within the nucleus.


Subject(s)
Thyroid Hormone Receptors beta/genetics , Adult , Animals , Cells, Cultured , Cloning, Molecular , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Pituitary Gland/chemistry , Pituitary Gland/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Thyroid Hormone Receptors beta/isolation & purification , Thyroid Hormone Receptors beta/metabolism
11.
Am J Chin Med ; 43(5): 879-92, 2015.
Article in English | MEDLINE | ID: mdl-26227398

ABSTRACT

Cinnamon is a traditional folk herb used in Asia and has been reported to have antidiabetic effects. Our previous study showed that cinnamaldehyde (CA), a major effective compound in cinnamon, exhibited hypoglycemic and hypolipidemic effects together in db/db mice. The aim of the present study was to elucidate the molecular mechanisms of the effects of CA on the transcriptional activities of three peroxisome proliferator-activated receptors, (PPAR) α, δ, and γ. We studied the effects of CA through a transient expression assay with TSA201 cells, derivatives of human embryonic kidney cell line (HEK293). Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis was also performed to evaluate mRNA expression levels. We show here that CA induced PPARδ, PPARγ and retinoid X receptor (RXR) activation. CA may activate PPARγ in a different manner than pioglitazone, as CA selectively stimulated PPARγ S342A mutant while pioglitazone did not. In addition, CA and L-165041 had a synergistic effect on PPARδ activation. To gather the biological evidence that CA increases PPARs transcription, we further measured the expressions of PPARδ and PPARγ target genes in 3T3-L1 adipocytes. The data showed CA induced the expression of PPARδ and PPARγ target genes, namely aP2 and CD36, in differentiated adipocytes. As a result, PPARδ, PPARγ and their heterodimeric partner RXR appear to play a part in the CA action in the target tissues, thereby enhancing insulin sensitivity and fatty acid ß-oxidation and energy uncoupling in skeletal muscle and adipose tissue.


Subject(s)
Acrolein/analogs & derivatives , Cinnamomum zeylanicum/chemistry , Gene Expression/drug effects , Insulin Resistance/genetics , PPAR delta/genetics , PPAR gamma/genetics , Retinoid X Receptors/genetics , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Acrolein/isolation & purification , Acrolein/pharmacology , Adipocytes/metabolism , Drug Synergism , Energy Metabolism/drug effects , Fatty Acids/metabolism , HEK293 Cells , Humans , Muscle, Skeletal/metabolism , Oxidation-Reduction/drug effects , PPAR delta/metabolism , PPAR gamma/metabolism , Phenoxyacetates/pharmacology , Pioglitazone , RNA, Messenger/genetics , Retinoid X Receptors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical , Thiazolidinediones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...