Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Bioenerg Biomembr ; 53(6): 679-692, 2021 12.
Article in English | MEDLINE | ID: mdl-34718921

ABSTRACT

The incidence of obesity has increased rapidly, becoming a worldwide public health issue that involves insulin resistance. A growing number of recent studies have demonstrated that microRNAs play a significant role in controlling the insulin signaling network. For example, miR-506-3p expression has been demonstrated to correlate with insulin sensitivity; however, the underlying mechanism remains unknown. In this study, we found that miR-506-3p enhanced glucose uptake by 2-deoxy-D-glucose uptake assays and regulated the protein expression of key genes involved in the PI3K/AKT insulin signaling pathway including IRS1, PI3K, AKT, and GlUT4. We next predicted ribosomal protein S6 kinase B1 (S6K1) to be a candidate target of miR-506-3p by bioinformatics analysis and confirmed using dual-luciferase assays that miR-506-3p regulated S6K1 expression by binding to its 3'-UTR. Moreover, modulating S6K1 expression counteracted the effects of miR-506-3p on glucose uptake and PI3K/AKT pathway activation. In conclusion, miR-506-3p altered IR in adipocytes by regulating S6K1-mediated PI3K/AKT pathway activation. Taken together, these findings provide novel insights and potential targets for IR therapy.


Subject(s)
Insulin Resistance , MicroRNAs , Adipocytes/metabolism , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction
2.
IUBMB Life ; 72(7): 1460-1467, 2020 07.
Article in English | MEDLINE | ID: mdl-32150331

ABSTRACT

Obesity is considered as a high-risk susceptibility state for most metabolic disorders and is directly related to preadipocyte differentiation or adipogenesis. Long noncoding RNAs (lncRNAs) are the key factors which have regulatory functions on various critical physiological and biological processes. PVT1 was identified as an oncogenic lncRNA which could promote angiogenesis in gastric cancer. However, the functions and molecular pathways related to PVT1 in adipogenesis had not been clarified yet. In the current study, the purpose was to identify the effects of lncRNA PVT1 on adipogenesis and the relevant molecular processes. Quantitative real-time polymerase chain reaction (RT-qPCR) was used to quantify PVT1 expression. The mechanism for PVT1 to participate in 3T3-L1 adipogenesis was identified by lentivirus-mediated gain- and loss-of-function tests. The potential association of PVT1 with cell viability was checked by CCK-8 assay and EdU staining. The gene expression for cytokines was determined by quantitative PCR (qPCR) and western blotting. PVT1 expression level was strongly upregulated after 3T3-L1 preadipocytes differentiated. In mice, PVT1 was abundantly expressed in adipose tissue, and the obese mice had higher PVT1 expression in adipose tissue than that of nonobese mice. Predominantly, PVT1 was found inside the nuclei. Overexpressed PVT1 could promote 3T3-L1 adipocyte differentiation as proved, which was the cause for the ability to accelerate lipid accumulation, by upregulating the expression of peroxisome proliferator activated receptor gamma, CCAAT/enhancer-binding protein α, and adipocyte protein 2, while knockdown of PVT1 caused opposite effects. The RNA immunoprecipitation demonstrated the binding relationship between PVT1 and STAT3 suggesting the potential role of STAT3 in 3T3-L1 preadipocyte differentiation. Furthermore, PVT1 could promote fatty acid synthesis but inhibit fatty acid oxidation. PVT1 was positively associated with 3T3-L1 preadipocyte differentiation, which highlighted the potential of PVT1 as a therapeutic target for obesity treatment.


Subject(s)
Adipocytes/cytology , Adipogenesis , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Cell Proliferation , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Survival , Male , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/metabolism , STAT3 Transcription Factor/genetics
3.
Exp Biol Med (Maywood) ; 239(12): 1567-74, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24903160

ABSTRACT

We previously proposed that LYR motif containing 1 (LYRM1)-induced mitochondrial reactive oxygen species (ROS) production contributes to obesity-related insulin resistance. Metformin inhibits ROS production and promotes mitochondrial biogenesis in specific tissues. We assessed the effects of metformin on insulin resistance in LYRM1-over-expressing 3T3-L1 adipocytes. Metformin enhanced basal and insulin-stimulated glucose uptake and GLUT4 translocation, reduced IRS-1 and Akt phosphorylation and ROS levels, and affected the expression of regulators of mitochondrial biogenesis in LYRM1-over-expressing adipocytes. Metformin may ameliorate LYRM1-induced insulin resistance and mitochondrial dysfunction in part via a direct antioxidant effect and in part by activating the adenosine monophosphate-activated protein kinase (AMPK)-PGC1/NRFs pathway.


Subject(s)
Adipocytes/drug effects , Adipocytes/physiology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Hypoglycemic Agents/metabolism , Insulin Resistance , Metformin/metabolism , Mitochondria/drug effects , Animals , Antioxidants/metabolism , Apoptosis Regulatory Proteins/metabolism , Cell Line , Mice , Reactive Oxygen Species/analysis
4.
Cell Biochem Biophys ; 70(1): 667-75, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24771405

ABSTRACT

LYR motif-containing 1 (LYRM1) was recently discovered to be involved in adipose tissue homeostasis and obesity-associated insulin resistance. We previously demonstrated that LYRM1 overexpression might contribute to insulin resistance and mitochondrial dysfunction. Additionally, knockdown of LYRM1 enhanced insulin sensitivity and mitochondrial function in 3T3-L1 adipocytes. We investigated whether knockdown of LYRM1 in 3T3-L1 adipocytes could rescue insulin resistance and mitochondrial dysfunction induced by the cyanide p-trifluoromethoxyphenyl-hydrazone (FCCP), a mitochondrion uncoupler, to further ascertain the mechanism by which LYRM1 is involved in obesity-associated insulin resistance. Incubation of 3T3-L1 adipocytes with 1 µM FCCP for 12 h decreased insulin-stimulated glucose uptake, reduced intracellular ATP synthesis, increased intracellular reactive oxygen species (ROS) production, impaired insulin-stimulated Glucose transporter type 4 (GLUT4) translocation, and diminished insulin-stimulated tyrosine phosphorylation of Insulin receptor substrate-1 (IRS-1) and serine phosphorylation of Protein Kinase B (Akt). Knockdown of LYRM1 restored insulin-stimulated glucose uptake, rescued intracellular ATP synthesis, reduced intracellular ROS production, restored insulin-stimulated GLUT4 translocation, and rescued insulin-stimulated tyrosine phosphorylation of IRS-1 and serine phosphorylation of Akt in FCCP-treated 3T3-L1 adipocytes. This study indicates that FCCP-induced mitochondrial dysfunction and insulin resistance are ameliorated by knockdown of LYRM1.


Subject(s)
Adipocytes/cytology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Gene Knockdown Techniques , Insulin Resistance/genetics , Mitochondria/drug effects , 3T3-L1 Cells , Adenosine Triphosphate/biosynthesis , Animals , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Proton Ionophores/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
5.
J Bioenerg Biomembr ; 44(6): 665-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22968630

ABSTRACT

NYGGF4 (also called PID1) is a recently discovered gene that is involved in obesity-related insulin resistance (IR). We aimed in the present study to further elucidate the effects of NYGGF4 on IR and the underlying mechanisms through using metformin treatment in 3T3-L1 adipocytes. Our data showed that the metformin pretreatment strikingly enhanced insulin-stimulated glucose uptake through increasing GLUT4 translocation to the PM in NYGGF4 overexpression adipocytes. NYGGF4 overexpression resulted in significant inhibition of tyrosine phosphorylation of IRS-1 and serine phosphorylation of Akt, whereas incubation with metformin strongly activated IRS-1 and Akt phosphorylation in NYGGF4 overexpression adipocytes. The reactive oxygen species (ROS) levels in NYGGF4 overexpression adipocytes were strikingly enhanced, which could be decreased by the metformin pretreatment. Our data also showed that metformin increased the expressions of PGC1-α, NRF-1, and TFAM, which were reduced in the NYGGF4 overexpression adipocytes. These results suggest that NYGGF4 plays a role in IR and its effects on IR could be reversed by metformin through activating IRS-1/PI3K/Akt and AMPK-PGC1-α pathways.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/metabolism , Hypoglycemic Agents/pharmacology , Insulin Resistance , Metformin/pharmacology , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/pathology , Animals , Carrier Proteins/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mice , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Transport/drug effects , Protein Transport/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors
6.
J Bioenerg Biomembr ; 44(5): 579-86, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22820890

ABSTRACT

Overexpression of the Homo sapiens LYR motif containing 1 (LYRM1) causes mitochondrial dysfunction and induces insulin resistance in 3T3-L1 adipocytes. α-Lipoic acid (α-LA), a dithiol compound with antioxidant properties, improves glucose transport and utilization in 3T3-L1 adipocytes. The aim of this study was to investigate the direct effects of α-LA on reactive oxygen species (ROS) production and insulin sensitivity in LYRM1 overexpressing 3T3-L1 adipocytes and to explore the underlying mechanism. Pretreatment with α-LA significantly increased both basal and insulin-stimulated glucose uptake and insulin-stimulated GLUT4 translocation, while intracellular ROS levels in LYRM1 overexpressing 3T3-L1 adipocytes were decreased. These changes were accompanied by a marked upregulation in expression of insulin-stimulated tyrosine phosphorylation of IRS-1 and serine phosphorylation of Akt following treatment with α-LA. These results indicated that α-LA protects 3T3-L1 adipocytes from LYRM1-induced insulin resistance partially via its capacity to restore mitochondrial function and/or increase phosphorylation of IRS-1 and Akt.


Subject(s)
Antioxidants/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Glucose/metabolism , Insulin Receptor Substrate Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Thioctic Acid/pharmacology , 3T3-L1 Cells , Animals , Apoptosis Regulatory Proteins/genetics , Gene Expression , Glucose/genetics , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Resistance/genetics , Mice , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Transport/drug effects , Protein Transport/genetics , Proto-Oncogene Proteins c-akt/genetics , Reactive Oxygen Species , Signal Transduction/genetics
7.
J Bioenerg Biomembr ; 44(3): 357-63, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22528396

ABSTRACT

NYGGF4 (also called PID1) was demonstrated that it may be related to the development of obesity-related IR. We aimed in the present study to further elucidate the effects of NYGGF4 on IR and the underlying mechanisms through using α-Lipoic acid (LA) treatment, which could facilitate glucose transport and utilization in fully differentiated adipocytes. Our data showed that the LA pretreatment strikingly enhanced insulin-stimulated glucose uptake through increasing GLUT4 translocation to the PM in NYGGF4 overexpression adipocytes. The reactive oxygen species (ROS) levels in NYGGF4 overexpression adipocytes were strikingly enhanced, which could be decreased by the LA pretreatment. NYGGF4 overexpression resulted in significant inhibition of tyrosine phosphorylation of IRS-1 and serine phosphorylation of Akt, whereas incubation with LA strongly activated IRS-1 and Akt phosphorylation in NYGGF4 overexpression adipocytes. These results suggest that LA protects 3T3-L1 adipocytes from NYGGF4-induced IR partially through increasing phosphorylation of IRS-1 and Akt and provide evidence that NYGGF4 may be a potential target for the treatment of obesity and obesity-related IR.


Subject(s)
Adipocytes/drug effects , Carrier Proteins/biosynthesis , Insulin Resistance/physiology , Proto-Oncogene Proteins c-akt/metabolism , Thioctic Acid/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/metabolism , Mice , Phosphorylation , Reactive Oxygen Species/metabolism , Transfection
8.
Exp Diabetes Res ; 2012: 820989, 2012.
Article in English | MEDLINE | ID: mdl-22110480

ABSTRACT

LYR motif containing 1 (LYRM1) is a novel gene that is abundantly expressed in the adipose tissue of obese subjects and is involved in insulin resistance. In this study, free fatty acids (FFAs) and tumor necrosis factor-α (TNF-α) are shown to upregulate LYRM1 mRNA expression in 3T3-L1 adipocytes. Conversely, resistin and rosiglitazone exert an inhibitory effect on LYRM1 mRNA expression. These results suggest that the expression of LYRM1 mRNA is affected by a variety of factors that are related to insulin sensitivity. LYRM1 may be an important mediator in the development of obesity-related insulin resistance.


Subject(s)
Adipokines/pharmacology , Apoptosis Regulatory Proteins/genetics , Fatty Acids, Nonesterified/pharmacology , Gene Expression Regulation/drug effects , Thiazolidinediones/pharmacology , 3T3-L1 Cells , Animals , Insulin Resistance/genetics , Mice , RNA, Messenger/analysis , Resistin/pharmacology , Rosiglitazone , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...