Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Gene ; 927: 148604, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38838872

ABSTRACT

OBJECTIVE: To extract exosomes from obese and non-obese mice, screen specifically expressed microRNAs by high-throughput sequencing and explore their roles. METHODS: An animal obesity model was constructed, and the successful construction of the obesity model was verified by HE staining, Western Blot and RT-qPCR. In addition, exosomes were extracted and verified by Western Blot. High-throughput sequencing was performed on the extracted serum exosomes to screen for differentially expressed microRNAs. fluorescence quantitative RT-PCR (RT-qPCR) was used to validate the differentially expressed miRNAs and explore their functions. RESULTS: 8 microRNAs were up-regulated and 11 microRNAs were down-regulated. mmu-miR-674-5p and X_28316 were significantly down-regulated and had the greatest impact on protein pathways. 8_13258 was significantly up-regulated and affected multiple protein pathways. GO enrichment analysis suggested that the differentially expressed microRNAs were mainly involved in the cleavage of microtubule activity, transferase activity/transferase pentameric acid. GO enrichment analysis suggested that differentially expressed microRNAs were mainly involved in the processes of cleavage microtubule activity, transferase activity/transfer pentamer, and threonine phosphatase/threonine kinase activity.KEGG pathway enrichment analysis showed that differentially expressed microRNAs were mainly involved in the processes of regulating the phosphorylation of TP53 activity, the G2/M DNA damage checkpoint, and the processing of the ends of DNA double-strand breaks. Protein interaction networks were enriched for Stat3, Fgr, Camk2b, Rac1, Asb6, and Ankfy1. Suggesting that they may be mediated by differential genes to participate in the process of insulin resistance. qRT-PCR results showed that the expression trend of mmu-miR-674-5p was consistent with the sequencing results. It suggests that it may be able to participate in the regulation of insulin resistance as a target gene. CONCLUSION: microRNAs were differentially expressed in serum exosomes of obese and non-obese mice and might be involved in the specific regulation of insulin resistance. mmu-miR-674-5p was differentially expressed significantly and the validation trend was consistent with it, suggesting that it might be able to participate in the regulation of insulin resistance as a target gene.

2.
Adipocyte ; 10(1): 350-360, 2021 12.
Article in English | MEDLINE | ID: mdl-34224297

ABSTRACT

Adipogenesis is regulated by genetic interactions, in which post-transcriptional regulation plays an important role. Staufen double-stranded RNA binding protein 1 (Staufen1 or STAU1) plays diverse roles in RNA processing and adipogenesis. Previously, we found that the downregulation of STAU1 affects the expression of fatty acid-binding protein 4 (FABP4) at the protein level but not at the mRNA level. This study aimed to determine the mechanism underlying the regulation of FABP4 expression by STAU1, explaining the inconsistency between FABP4 mRNA and protein levels. We used RNA interference, photoactivatable ribonucleoside enhanced cross-linking and immunoprecipitation, and an adeno-associated virus to examine the functions of STAU1 in adipogenesis. Our results indicate that STAU1 binds to the coding sequences of FABP4, thereby regulating the translation of FABP4 mRNA by unwinding the double-stranded structure. Furthermore, STAU1 mediates adipogenesis by regulating the secretion of free fatty acids. However, STAU1 knockdown decreases the fat weight/body weight ratio but does not affect the plasma triglyceride levels. These findings describe the mechanisms involved in STAU1-mediated regulation of FABP4 expression at the translational level during adipogenesis.


Subject(s)
Adipogenesis , Fatty Acid-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Adipogenesis/genetics , Animals , Cytoskeletal Proteins/metabolism , Mice , Protein Biosynthesis , RNA, Messenger/genetics
3.
Aging (Albany NY) ; 13(11): 15164-15192, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34081618

ABSTRACT

Long non-coding RNAs (lncRNAs) comprise an integral part of the eukaryotic transcriptome. Alongside proteins, lncRNAs modulate lncRNA-based gene signatures of unstable transcripts, play a crucial role as antisense lncRNAs to control intracellular homeostasis and are implicated in tumorigenesis. However, the role of genomic instability-associated lncRNAs in low-grade gliomas (LGG) has not been fully explored. In this study, lncRNAs expression and somatic mutation profiles in low-grade glioma genome were used to identify eight novel mutant-derived genomic instability-associated lncRNAs including H19, FLG-AS1, AC091932.1, AC064875.1, AL138767.3, AC010273.2, AC131097.4 and ISX-AS1. Patients from the LGG gene mutagenome atlas were grouped into training and validation sets to test the performance of the signature. The genomic instability-associated lncRNAs signature (GILncSig) was then validated using multiple external cohorts. A total of 59 novel genomic instability-associated lncRNAs in LGG were used for least absolute shrinkage and selection operator (Lasso), single and multifactor Cox regression analysis using the training set. Furthermore, the independent predictive role of risk features in the training and validation sets were evaluated through survival analysis, receiver operating feature analysis and construction of a nomogram. Patients with IDH1 mutation status were grouped into two different risk groups based on the GILncSig score. The low-risk group showed a relatively higher rate of IDH1 mutations compared with patients in the high-risk group. Furthermore, patients in the low-risk group had better prognosis compared with patients in the high-risk group. In summary, this study reports a reliable prognostic prediction signature and provides a basis for further investigation of the role of lncRNAs on genomic instability. In addition, lncRNAs in the signature can be used as new targets for treatment of LGG.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Gene Expression Profiling , Genomic Instability , Glioma/genetics , Glioma/pathology , RNA, Long Noncoding/genetics , Adult , Area Under Curve , Female , Filaggrin Proteins , Gene Expression Regulation, Neoplastic , Humans , Isocitrate Dehydrogenase/genetics , Kaplan-Meier Estimate , Male , Multivariate Analysis , Mutation/genetics , Neoplasm Grading , Prognosis , Proportional Hazards Models , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , ROC Curve , Reproducibility of Results
4.
Life Sci ; 265: 118762, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33189825

ABSTRACT

AIMS: This study is to investigate the role of adenovirus type 36 (Ad36) in inducing differentiation of human adipose-derived stem cells (hADSCs) into brown adipocytes. MAIN METHODS: The hADSCs were induced to differentiate into adipocytes by a cocktail method and Ad36, respectively. They were collected on the 2nd, 4th, 6th, and 8th day, respectively. LncRNA ROR was silenced by siRNA. RT-qPCR and Western-blot were used to detect the mRNA and protein levels. Transmission electron microscopy was used to observe the mitochondria. KEY FINDINGS: The mRNA and protein expression levels of LncRNA ROR, Cidea, Dio2, Fgf21, Ucp1, Prdm16, Cox5b, Atp5o, Atp6, and Nd2 in the Ad36 induction group were significantly higher than those in the cocktail induction group. The expression levels of Leptin mRNA and protein in the Ad36 induction group were significantly lower than those in the cocktail induction group. After siRNA knockdown of LncRNA ROR, mRNA and protein expression levels of Cidea, Dio2, Fgf21, Ucp1, Prdm16, Cox5b, Atp5o, Atp6 and Nd2 were significantly lower than the control group during the induction of hADSC differentiation into adipocytes by Ad36. Additionally, mitochondria in the Ad36 induction group was increased compared to that in the cocktail induction group. SIGNIFICANCE: Ad36 may promote the differentiation of hADSCs into brown adipocytes by up-regulating LncRNA ROR.


Subject(s)
Adenoviridae/metabolism , Adenovirus Infections, Human/metabolism , Adipocytes, Brown/virology , Mesenchymal Stem Cells/metabolism , RNA, Long Noncoding/metabolism , Adipocytes, Brown/metabolism , Adipocytes, Brown/physiology , Adipocytes, Brown/ultrastructure , Blotting, Western , Cell Differentiation , Gene Expression Regulation , Gene Silencing , Humans , Microscopy, Electron, Transmission , Mitochondria/metabolism , Real-Time Polymerase Chain Reaction
5.
Lipids Health Dis ; 18(1): 70, 2019 Mar 21.
Article in English | MEDLINE | ID: mdl-30902099

ABSTRACT

BACKGROUND: This study aims to investigate the molecular mechanism of Adenovirus type 36 (Ad36) in adipocyte differentiation and glucolipid metabolism. METHODS: Rat obesity model was established by Ad36 infection and high-fat diet, respectively. Comparison of the body weight, clinical biochemical indicators, insulin sensitivity and lipid heterotopic deposition between these two models was performed. Ad36-induced adipocyte in vitro model was also established. The binding rate of FoxO1, PPARγ and its target gene promoter was detected using ChIP. The mRNA and protein expression levels of PPARγ and downstream target genes were detected by RT-PCR and Western blot, respectively. Oil red O staining was used to measure differentiation into adipocyte. Wortmannin (WM), inhibitor of PI3K, was used to act on Ad36-induced hADSCs. RESULTS: Ad36-induced obese rats did not exhibit disorders in blood glucose and blood TG, insulin resistance and lipid ectopic deposition. The expression of Adipoq, Lpin1 and Glut4 in the adipose tissue increased. Oil red O staining showed that Ad36 induced the differentiation of hAMSCs into human adipocytes in vitro. During this process, the binding rate of FoxO1 and PPARγ promoter regions was weakened. However, the binding rate of the transcription factor PPARγ to its target genes Acc, Adipoq, Lpin1 and Glut4 was enhanced, and thus increased the protein expression of P-FoxO1, PPARγ2, ACC, LPIN1, GLUT4 and ADIPOQ. The PI3K inhibitor Wortmannin reduced the expression of P-Akt, P-FoxO1 and PPARγ2, thereby inhibiting adipogenesis of hADSC. CONCLUSION: Ad36 may promote fatty acid and triglyceride synthesis, and improve insulin sensitivity by affecting the PI3K/Akt/FoxO1/PPARγ signaling pathway.


Subject(s)
Adipose Tissue/metabolism , Obesity/genetics , PPAR gamma/genetics , Stem Cells/cytology , Adipocytes/metabolism , Adipocytes/virology , Adiponectin/genetics , Adipose Tissue/cytology , Adipose Tissue/virology , Animals , Cell Differentiation/genetics , Diet, High-Fat/adverse effects , Forkhead Box Protein O1/genetics , Gene Expression Regulation, Developmental , Glucose/metabolism , Glucose Transporter Type 4/genetics , Humans , Lipid Metabolism/genetics , Obesity/metabolism , Obesity/pathology , Obesity/virology , Phosphatidate Phosphatase/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction/drug effects , Stem Cells/virology , Wortmannin/administration & dosage
6.
Biochem Biophys Res Commun ; 495(2): 1878-1884, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29217197

ABSTRACT

AIMS: To determine the role of lncRNA HOXA11-AS1 on adipocyte differentiation. METHODS: Human adipose-derived stem cells (hADSCs) were isolated from adipose tissues of patients and cultured in vitro, followed by knockdown of HOXA11-AS1. Then, adipocyte differentiation and expression of adipogenic-related genes (CEBP-α, DGAT2, CIDEC, and perilipin) were measured by RT-qPCR and Western blot. RESULTS: We demonstrated that knockdown of HOXA11-AS1 inhibited adipocyte differentiation, leading to suppression of adipogenic-related gene (CEBP-α, DGAT2, CIDEC, and perilipin) transcription, as well as decreased lipid accumulation in hADSCs. In addition, lncRNA HOXA11-AS1 was highly expressed in obese patients and significantly increased during the process of adipocyte differentiation. CONCLUSION: The results provide new insight into the molecular mechanism by which lncRNA HOXA11-AS1 is involved in adipogenesis and may have implications for the treatment of obesity and associated disorders.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Adipose Tissue/growth & development , Homeodomain Proteins/metabolism , Obesity/metabolism , RNA, Long Noncoding/metabolism , Stem Cells/physiology , Adipocytes/pathology , Adipose Tissue/pathology , Cell Differentiation/physiology , Cells, Cultured , Humans , Stem Cells/pathology
7.
Mol Cell Biochem ; 428(1-2): 1-8, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27995415

ABSTRACT

This study is to investigate the role of adenovirus 36 (Ad36) in regulating expression of peroxisome proliferator-activated receptor γ (PPARγ) and cell death-inducing DFFA-like effector c (CIDEC) in Ad36-induced adipocyte differentiation. Human adipose-derived mesenchymal stem cells (hAMSCs) were isolated and cultured, and then infected with Ad36. Ad36-induced adipocytes were identified using quantitative real-time PCR and Oil red O staining. The expression levels of PPARγ and CIDEC in Ad36-induced adipocytes were determined by quantitative real-time PCR and Western blot analysis. Glucose uptake and intracellular triglyceride content were also determined in these induced cells. Our results from the Oil red O staining showed that Ad36 induced the differentiation of hAMSCs into human adipocytes in vitro. Moreover, the medium glucose concentration was significantly decreased, while the intracellular triglyceride content was significantly increased, in the Ad36-induced adipocytes, compared with the control group. Furthermore, our results showed that, the mRNA and protein expression levels of PPARγ and CIDEC were significantly upregulated in Ad36-induced adipocytes, in a time-dependent manner. On the other hand, compared with the control group, the CIDEC expression was downregulated when the Ad36-induced adipocytes were treated with the PPARγ inhibitor, GW9662. Ad36 could upregulate the expression level of CIDEC through increasing PPARγ expression during the adipocyte differentiation process.


Subject(s)
Adenoviridae/physiology , Adipocytes/cytology , Mesenchymal Stem Cells/virology , PPAR gamma/genetics , Proteins/genetics , Adipocytes/metabolism , Adipocytes/virology , Anilides/pharmacology , Apoptosis Regulatory Proteins , Cell Differentiation , Cells, Cultured , Glucose/metabolism , Humans , Mesenchymal Stem Cells/cytology , PPAR gamma/metabolism , Proteins/metabolism , Time Factors , Triglycerides/metabolism , Up-Regulation/drug effects
8.
Diagn Pathol ; 9: 83, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24739504

ABSTRACT

OBJECTIVE: This study is to determine if Adenovirus type 36 (Ad36) infection is related to macrophage infiltration in the obese group and non-obese group and the related molecular mechanisms. METHODS: Ninety obesity patients and 95 non-obesity Uygur individuals were enrolled in this study. CD68 levels in abdominal subcutaneous and omental adipose tissues were detected by immunohistochemistry. The cytokine expression levels of adiponectin (APMI) and visfatin in serum were measured by enzyme-linked immunosorbent assay. Infection of 3T3-L1 cells with Ad36 was performed. Real-time PCR was performed to determine expression levels of APMI and Visfatin genes in the 3T3-L1 preadipocytes infected with Ad36. RESULTS: In the obese individuals infected with Ad36, the expression levels of adiponectin and visfatin in serum was elevated. For the individuals infected with Ad36, the macrophage infiltration (as indicated by CD68 level) in the obese group was also significantly higher than that in the non-obese group (P < 0.05) in both abdominal subcutaneous and omental adipose tissues. The real-time PCR results indicated that APMI mRNA levels and Visfatin mRNA levels in Ad36 infected cells were significantly increased. CONCLUSIONS: Ad36 infection may be a factor related with macrophage infiltration in adipose tissues of the obese patients. The APMI and Visfatin genes may be involved in the mechanism underlying the effect of Ad36 infection on the obese patients. VIRTUAL SLIDES: The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1849614638119816.


Subject(s)
Adenovirus Infections, Human/blood , Adiponectin/blood , Cytokines/blood , Nicotinamide Phosphoribosyltransferase/blood , Overweight/blood , 3T3-L1 Cells , Abdominal Fat/metabolism , Adenovirus Infections, Human/epidemiology , Adenovirus Infections, Human/genetics , Adipocytes/metabolism , Adipocytes/virology , Adiponectin/genetics , Adiponectin/metabolism , Adult , Aged , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Case-Control Studies , China/epidemiology , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Macrophages/metabolism , Mice , Middle Aged , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Overweight/epidemiology , Overweight/genetics , Time Factors
9.
Int J Clin Exp Med ; 7(11): 4322-6, 2014.
Article in English | MEDLINE | ID: mdl-25550948

ABSTRACT

OBJECTIVES: This study is to determine if two adipocytokines, adiponectin and visfatin, can be used as diagnosis markers for metabolic syndrome (MS) in Uygur population. METHODS: Sixty-two MS patients and 41 control individuals with normal body weights were enrolled in this study. Abdominal subcutaneous and omental adipose tissues were collected for determination of biochemical indices. The adipokines serum levels were determined by enzyme-linked immunosorbent assay (ELISA). Blood were collected from the MS patients and the control individuals and extracted proteins and RNAs subjected to western blot analysis and real-time PCR to determine adiponectin and visfatin expression, respectively. RESULTS: ELISA indicated that the serum adiponectin in the MS group was decreased (0.59 ± 0.21 versus 0.49 ± 0.18) in comparison with the control group (P < 0.05). But the serum visfatin in the MS group were increased (1.07 ± 0.41 versus 1.25 ± 0.32) when compared with the control group (P < 0.05). The western blot revealed decreased adiponectin and increased visfatin expression in the MS patients when compared with the normal controls. Further real-time RT-PCR analysis showed that the adiponectin and visfatin expression are altered via a transcriptional mechanism. CONCLUSIONS: Adiponectin and visfatin might be used as diagnosis markers of MS in Uygur population.

SELECTION OF CITATIONS
SEARCH DETAIL
...