Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Front Immunol ; 14: 1232187, 2023.
Article in English | MEDLINE | ID: mdl-38090557

ABSTRACT

Introduction: Food allergies have a substantial impact on patient health, but their mechanisms are poorly understood, and strategies for diagnosing, preventing, and treating food allergies are not optimal. This study explored the levels of and relationship between IL-17A and IFN-γ in the saliva of children with food allergies, which will form the basis for further mechanistic discoveries as well as prevention and treatment measures for food allergies. Methods: A case-control study with 1:1 matching was designed. Based on the inclusion criteria, 20 case-control pairs were selected from patients at the Skin and Allergy Clinic and children of employees. IL-17A and IFN-γ levels in saliva were measured with a Luminex 200 instrument. A general linear model was used to analyze whether the salivary IL-17A and IFN-γ levels in the food allergy group differed from those in the control group. Results: The general linear model showed a significant main effect of group (allergy vs. healthy) on the levels of IL-17A and IFN-γ. The mean IL-17A level (0.97 ± 0.09 pg/ml) in the food allergy group was higher than that in the healthy group (0.69 ± 0.09 pg/ml). The mean IFN-γ level (3.0 ± 0.43 pg/ml) in the food allergy group was significantly higher than that in the healthy group (1.38 ± 0.43 pg/ml). IL-17A levels were significantly positively related to IFN-γ levels in children with food allergies (r=0.79) and in healthy children (r=0.98). Discussion: The salivary IL-17A and IFN-γ levels in children with food allergies were higher than those in healthy children. This finding provides a basis for research on new methods of diagnosing food allergies and measuring the effectiveness of treatment.


Subject(s)
Food Hypersensitivity , Interferon-gamma , Child , Humans , Interleukin-17 , Case-Control Studies , Skin , Food Hypersensitivity/diagnosis
2.
BMC Endocr Disord ; 23(1): 238, 2023 Oct 30.
Article in English | MEDLINE | ID: mdl-37904219

ABSTRACT

BACKGROUND: The prevalence of childhood obesity and overweight has risen globally, leading to increased rates of metabolic disorders. Various factors, including genetic, epigenetic, and environmental influences such as diet and physical activity, contribute to pediatric obesity. This study aimed to identify specific circulating miRNAs as potential biomarkers for assessing obesity in children. METHODS: Thirty children, including 15 obese and 15 extremely thin individuals, were selected for this study. MiRNA expression in circulating plasma was assessed using miRNA microarrays. The reliability of differential miRNA expression was confirmed using TaqMan qPCR. The correlation between miRNAs and obesity was analyzed through multiple linear regression, receiver operator characteristic (ROC) curve analysis, and odds ratio (OR) calculations. Bioinformatics tools were utilized to identify target genes for the selected miRNAs, and a functional network map was constructed. RESULTS: A total of 36 differentially expressed miRNAs were identified through gene chip analysis, and TaqMan qPCR validation confirmed the upregulation of seven miRNAs: hsa-miR-126-3p, hsa-miR-15b-5p, hsa-miR-199a-3p, hsa-miR-20a-5p, hsa-miR-223-3p, hsa-miR-23a-3p, and hsa-miR-24-3p. Among these, hsa-miR-15b-5p and hsa-miR-223-3p exhibited a statistically significant difference except for hsa-miR-23a-3p. These two miRNAs showed more predicted target genes related to obesity than others. Multiple linear regression analysis revealed an association between obesity and hsa-miR-15b-5p and hsa-miR-223-3p [10.529 (4.974-16.084), -10.225 (-17.852~ -2.657)]. Even after adjusting for age and sex, these two miRNAs remained associated with obesity [8.936 (3.572-14.301), -8.449(-15.634~ -1.303)]. The area under the ROC curve (AUC) reached values of 0.816, 0.711, and 0.929, respectively. Odds ratio analysis demonstrated a significant correlation between obesity and hsa-miR-15b-5p (OR = 143, 95% CI 5.80 to 56,313, p = 0.024) and between obesity and hsa-miR-223-3p (OR = 0.01, 95% CI 0.00 to 0.23, p = 0.037). Importantly, hsa-miR-15b-5p was found to have numerous target genes associated with the FoxO, insulin, Ras, and AMPK signaling pathways. CONCLUSIONS: Differential miRNA expression profiles in the circulation of obese children compared to controls suggest underlying metabolic abnormalities. Hsa-miR-15b-5p and hsa-miR-223-3p may be considered as molecular markers for the screening of obese children and populations at risk of developing metabolic syndrome.


Subject(s)
MicroRNAs , Pediatric Obesity , Child , Humans , Pediatric Obesity/diagnosis , Pediatric Obesity/epidemiology , Pediatric Obesity/genetics , Reproducibility of Results , MicroRNAs/metabolism , Biomarkers , Signal Transduction/genetics
3.
Front Cell Dev Biol ; 10: 954769, 2022.
Article in English | MEDLINE | ID: mdl-36120562

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) shows complicated and not clearly defined etiology and pathogenesis. Although no pharmacotherapeutics have improved the survival rate in HFpEF, exercise training has become an efficient intervention to improve functional outcomes. Here, we investigated N6-methyladenosine (m6A) RNA methylation modification in a "two-hit" mouse model with HFpEF and HFpEF with exercise (HFpEF + EXT). The manner of m6A in HFpEF and HFpEF + EXT hearts was explored via m6A-specific methylated RNA immunoprecipitation followed by high-throughput and RNA sequencing methods. A total amount of 3992 novel m6A peaks were spotted in HFpEF + EXT, and 426 differently methylated sites, including 371 hypermethylated and 55 hypomethylated m6A sites, were singled out for further analysis (fold change >2, p < 0.05). According to gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, unique m6A-modified transcripts in HFpEF + EXT were associated with apoptosis-related pathway and myocardial energy metabolism. HFpEF + EXT had higher total m6A levels and downregulated fat mass and obesity-related (FTO) protein levels. Overexpression of FTO cancels out the benefits of exercise in HFpEF + EXT mice by promoting myocyte apoptosis, myocardial fibrosis and myocyte hypertrophy. Totally, m6A is a significant alternation of epitranscriptomic processes, which is also a potentially meaningful therapeutic target.

4.
Front Endocrinol (Lausanne) ; 13: 890305, 2022.
Article in English | MEDLINE | ID: mdl-35769075

ABSTRACT

Background: Serum uric acid (SUA) is associated with many cardiovascular risk factors, such as metabolic syndrome (MetS) and subclinical atherosclerosis. However, the relationship of SUA with carotid atherosclerosis remains controversial. We aimed to investigate whether elevated SUA levels are associated with a high risk of carotid atherosclerosis and MetS in patients with type 2 diabetes mellitus (T2DM). Methods: This cross-sectional study was performed with a sample of 1,947 hospitalized patients with T2DM. Carotid intima-media thickness and carotid artery plaques were measured via Doppler ultrasound. Results: Uric acid levels were negatively associated with HbA1C, eGFR, and HDL-C (all P < 0.001) and positively associated with WBC, BMI, ACR, creatinine, total cholesterol, triglycerides, LDL-C, systolic blood pressure, and diastolic blood pressure (all P < 0.001). After adjusting for multiple potential confounders, the risks were substantially higher for MetS in the highest quartile of SUA levels (odds ratio: 2.91, 95% confidence interval: 1.54-5.51, P = 0.003 for trend) than in the lowest quartile of SUA levels. Furthermore, a significant increase was observed in the prevalence of overweight/obesity, hypertension, and dyslipidemia across the SUA quartiles independent of confounders. However, no significant association was found between SUA quartile with the presence of carotid atherosclerosis. Conclusions: In patients with T2DM, SUA levels were closely associated with MetS and its components but not with carotid atherosclerosis.


Subject(s)
Carotid Artery Diseases , Diabetes Mellitus, Type 2 , Metabolic Syndrome , Carotid Artery Diseases/epidemiology , Carotid Artery Diseases/etiology , Carotid Intima-Media Thickness , Cross-Sectional Studies , Diabetes Mellitus, Type 2/complications , Humans , Metabolic Syndrome/complications , Metabolic Syndrome/epidemiology , Uric Acid
5.
Front Cell Dev Biol ; 9: 702579, 2021.
Article in English | MEDLINE | ID: mdl-34368154

ABSTRACT

In this study, we aimed to systematically profile global RNA N6-methyladenosine (m6A) modification patterns in a mouse model of diabetic cardiomyopathy (DCM). Patterns of m6A in DCM and normal hearts were analyzed via m6A-specific methylated RNA immunoprecipitation followed by high-throughput sequencing (MeRIP-seq) and RNA sequencing (RNA-seq). m6A-related mRNAs were validated by quantitative real-time PCR analysis of input and m6A immunoprecipitated RNA samples from DCM and normal hearts. A total of 973 new m6A peaks were detected in DCM samples and 984 differentially methylated sites were selected for further study, including 295 hypermethylated and 689 hypomethylated m6A sites (fold change (FC) > 1.5, P < 0.05). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway analyses indicated that unique m6A-modified transcripts in DCM were closely linked to cardiac fibrosis, myocardial hypertrophy, and myocardial energy metabolism. Total m6A levels were higher in DCM, while levels of the fat mass and obesity-associated (FTO) protein were downregulated. Overexpression of FTO in DCM model mice improved cardiac function by reducing myocardial fibrosis and myocyte hypertrophy. Overall, m6A modification patterns were altered in DCM, and modification of epitranscriptomic processes, such as m6A, is a potentially interesting therapeutic approach.

6.
BMC Cancer ; 19(1): 405, 2019 Apr 29.
Article in English | MEDLINE | ID: mdl-31035970

ABSTRACT

BACKGROUND: Wilms' tumor is also called nephroblastoma and is the most common pediatric renal cancer. Several genetic and epigenetic factors have been found to account for the development of Wilms' tumor. MiRNAs play important roles in this tumorigenic process. In the present study, we aimed to investigate the role of miR-140-5p in nephroblastoma by identifying its targets, as well as its underlying molecular mechanism of action. METHODS: The miRNA expression profile of nephroblastoma samples was investigated and the targets of miR-140-5p were predicted and validated using the miRNA luciferase reporter method. Moreover, the roles of miR-140-5p in regulating nephroblastoma cell proliferation, migration and cell cycle were analyzed by the CCK8, migration and flow cytometry assays, respectively. The downstream protein of the direct target of miR-140-5p was also identified. RESULTS: miR-140-5p was downregulated in Wilms' tumor tissues, whereas in the nephroblastoma cell lines G401 and WT-CLS1 that exhibited high levels of miRNA-140-5p, inhibition of cellular proliferation and metastasis were noted as well as cell cycle arrest at the G1/S phase. TGFBRI and IGF1R were identified as direct target genes for miRNA-140-5p. In addition, SMAD2/3 and p-AKT were regulated by TGFBRI and IGF1R separately and participated in the miRNA-140-5p regulatory network. Ectopic expression of TGFBR1 and IGF-1R could abrogate the inhibitory effect of miR-140-5p. CONCLUSION: We demonstrated that miRNA-140-5p participates in the progression of Wilms' tumor by targeting the TGFBRI/SMAD2/3 and the IGF-1R/AKT signaling pathways.


Subject(s)
Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Signal Transduction/genetics , Wilms Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Child , Disease Progression , Down-Regulation , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Profiling , Gene Regulatory Networks/genetics , Humans , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1 , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Wilms Tumor/metabolism , Wilms Tumor/pathology
7.
Biochem Biophys Res Commun ; 511(4): 855-861, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30850164

ABSTRACT

Adipogenesis is one of the key processes during obesity development. Better understanding of this process could advance our knowledge on obesity and its treatment. Transcription factors (TFs) are master regulators during adipogenesis, however, a system-wide analysis of TFs dynamic proteome during adipogenesis is lacking. Here, we profiled 472 TFs and systematically elucidated their roles during the first 7 days of adipogenesis of human adipose-derived stem cells (hADSCs) on proteome scale. We identified two main and four sub-phases during adipogenesis. The commitment phase (0 h-8 h) mainly mediated stem cell proliferation, differentiation and chromatin remodeling, in which proteins of SWI/SNF family are the key centroid nodes. The determination phase (1D-7D) predominately regulated fat cell differentiation and response to lipid and oxygen, which could be associated with terminal differentiation of adipocyte and responsible for maturation. PPARγ, CREB1 and MYC are the centroid nodes of this phase. Remarkably, we identified and verified three TFs (BATF3, MAFF and MXD4) as novel regulators of adipogenesis, whose over-expression could inhibit adipogenesis of hADSCs in vitro. Overall, our study provided a valuable TFs resource to understand the complex process of adipogenesis.


Subject(s)
Adipocytes/cytology , Adipogenesis , Adipose Tissue/cytology , Stem Cells/cytology , Transcription Factors/genetics , Adipocytes/metabolism , Adipose Tissue/metabolism , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Developmental , Humans , Obesity/genetics , Proteomics , Stem Cells/metabolism
8.
J Cell Physiol ; 234(6): 9475-9485, 2019 06.
Article in English | MEDLINE | ID: mdl-30362570

ABSTRACT

Macrophages and many chemokines are closely associated with the adipogenic differentiation of bone marrow mesenchymal stem cells (MSCs), but their roles in adipogenesis and the underlying mechanisms are not fully understood. Here, we first investigated the influence of macrophages on the differentiation of MSCs in vitro. We found that RAW246.7 macrophages cocultured with MSCs strongly blocked the differentiation progress and inhibited the expression of C-X-C motif chemokine ligand 1 (CXCL1) during adipogenesis. Coculture with MSCs mainly induced macrophages toward M2 polarization. In addition, the expression of CXCL1 and its receptor, C-X-C chemokine receptor type 2, CXCR2 are high during adipogenic differentiation of MSCs and not in mature adipocytes. Although CXCL1 had no effect on adipogenesis, treatment with a specific CXCR2 inhibitor, SB225002, hampered the adipogenic differentiation of MSCs. Blocking CXCR2 decreased p38 and Elk1 phosphorylation but increased the extracellular signal-regulated kinase (ERK) phosphorylation at the initial stage of adipogenesis, which suppressed the phosphorylation of p38/ERK-Elk1 at the late stage. Inhibition of ERK had similar effects on adipogenesis and Elk1 phosphorylation. Our data suggest that MSCs interact with macrophages during adipogenic differentiation. CXCR2 regulates the adipogenic differentiation of MSCs by altering the activation of the p38/ERK-Elk1 signaling pathway.


Subject(s)
Adipogenesis , Macrophages/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Receptors, Interleukin-8B/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Animals , Cell Polarity/drug effects , Chemokine CXCL1/pharmacology , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , ets-Domain Protein Elk-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Immunol Lett ; 202: 23-30, 2018 10.
Article in English | MEDLINE | ID: mdl-30076857

ABSTRACT

Vitamin D insufficiency is associated with chronic inflammatory diseases. However, the mechanism by which vitamin D reduces obesity-related inflammation remains poorly understood. In this study, we investigated the inhibitory effects of vitamin D on palmitate-induced inflammatory response in macrophages and explored the potential mechanisms of vitamin D action. The effect of vitamin D on the expression of inflammatory factors induced by palmitate, a saturated fatty acid, was investigated using human THP-1 macrophages and murine RAW 264.7 cells. To elucidate the mechanism by which vitamin D affects palmitate-induced inflammatory cytokine production, we investigated the activity of stress kinase-related proteins. Palmitate significantly increased TNF-α and IL-6 expression and secretion in THP-1 and RAW 264.7 macrophages. Treatment with the active form of vitamin D inhibited palmitate-induced TNF-α and IL-6 production in macrophages. Furthermore, vitamin D significantly reduced palmitate-stimulated activation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase 1/2 (ERK1/2). The mitogen-activated protein kinase signaling pathway partly accounts for the induction of pro-inflammatory cytokines by palmitate. Our data suggest that the attenuation of palmitate-induced TNF-α and IL-6 gene expression and protein secretion by vitamin D are associated with reduced activation of JNK and ERK1/2.


Subject(s)
Cytokines/metabolism , Inflammation/drug therapy , Macrophages/drug effects , Mitogen-Activated Protein Kinases/metabolism , Palmitates/pharmacology , Signal Transduction/drug effects , Vitamin D/pharmacology , Animals , Cell Line , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Macrophages/metabolism , Mice , RAW 264.7 Cells , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Nutr Biochem ; 59: 76-83, 2018 09.
Article in English | MEDLINE | ID: mdl-29986310

ABSTRACT

Folic acid (FA) supplementation may protect from obesity and insulin resistance, the effects and mechanism of FA on chronic high-fat-diet-induced obesity-related metabolic disorders are not well elucidated. We adopted a genome-wide approach to directly examine whether FA supplementation affects the DNA methylation profile of mouse adipose tissue and identify the functional consequences of these changes. Mice were fed a high-fat diet (HFD), normal diet (ND) or an HFD supplemented with folic acid (20 µg/ml in drinking water) for 10 weeks, epididymal fat was harvested, and genome-wide DNA methylation analyses were performed using methylated DNA immunoprecipitation sequencing (MeDIP-seq). Mice exposed to the HFD expanded their adipose mass, which was accompanied by a significant increase in circulating glucose and insulin levels. FA supplementation reduced the fat mass and serum glucose levels and improved insulin resistance in HFD-fed mice. MeDIP-seq revealed distribution of differentially methylated regions (DMRs) throughout the adipocyte genome, with more hypermethylated regions in HFD mice. Methylome profiling identified DMRs associated with 3787 annotated genes from HFD mice in response to FA supplementation. Pathway analyses showed novel DNA methylation changes in adipose genes associated with insulin secretion, pancreatic secretion and type 2 diabetes. The differential DNA methylation corresponded to changes in the adipose tissue gene expression of Adcy3 and Rapgef4 in mice exposed to a diet containing FA. FA supplementation improved insulin resistance, decreased the fat mass, and induced DNA methylation and gene expression changes in genes associated with obesity and insulin secretion in obese mice fed a HFD.


Subject(s)
Adipose Tissue/drug effects , DNA Methylation/drug effects , Diet, High-Fat/adverse effects , Folic Acid/pharmacology , Insulin Resistance/physiology , Adenylyl Cyclases/genetics , Adipose Tissue/physiology , Animals , Biomarkers/blood , Blood Glucose/metabolism , Dietary Supplements , Energy Metabolism/drug effects , Enzymes/genetics , Enzymes/metabolism , Guanine Nucleotide Exchange Factors/genetics , Male , Mice, Inbred C57BL , Obesity/genetics , Reproducibility of Results
11.
Nutr Metab (Lond) ; 14: 68, 2017.
Article in English | MEDLINE | ID: mdl-29118818

ABSTRACT

BACKGROUND: Folic acid (FA) is an antioxidant that can reduce reactive oxygen species generation and can blunt cardiac dysfunction during ischemia. We hypothesized that FA supplementation prevents cardiac fibrosis and cardiac dysfunction induced by obesity. METHODS: Six-week-old C57BL6/J mice were fed a high-fat diet (HFD), normal diet (ND), or an HFD supplemented with folic acid (FAD) for 14 weeks. Cardiac function was measured using a transthoracic echocardiographic exam. Phenotypic analysis included measurements of body and heart weight, blood glucose and tissue homocysteine (Hcy) content, and heart oxidative stress status. RESULTS: HFD consumption elevated fasting blood glucose levels and caused obesity and heart enlargement. FA supplementation in HFD-fed mice resulted in reduced fasting blood glucose, heart weight, and heart tissue Hcy content. We also observed a significant cardiac systolic dysfunction when mice were subjected to HFD feeding as indicated by a reduction in the left ventricular ejection fraction and fractional shortening. However, FAD treatment improved cardiac function. FA supplementation protected against cardiac fibrosis induced by HFD. In addition, HFD increased malondialdehyde concentration of the heart tissue and reduced the levels of antioxidant enzyme, glutathione, and catalase. HFD consumption induced myocardial oxidant stress with amelioration by FA treatment. CONCLUSION: FA supplementation significantly lowers blood glucose levels and heart tissue Hcy content and reverses cardiac dysfunction induced by HFD in mice. These functional improvements of the heart may be mediated by the alleviation of oxidative stress and myocardial fibrosis.

12.
Mol Med Rep ; 16(4): 3799-3806, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28765929

ABSTRACT

MicroRNAs (miRNAs) are implicated in the pathogenesis of obesity. The aim of the present study was to characterize the miRNA profile associated with early childhood obesity in peripheral blood mononuclear cells (PBMCs). A total of 12 children (6 obese and 6 lean controls) aged 36 months old to 48 months old were recruited. The miRNA expression profile from PBMCs was detected using the multiplexed NanoString nCounter system. Bioinformatics was employed to detect target genes and miRNA­regulated biological function. A total of 9 differentially expressed miRNAs were identified in obese children compared with lean children (P<0.05). Among the 9 miRNAs, miR­199a­3p/miR­199b­3p and miR­4454 presented at least a 1.5­fold change in expression. A total of 643 potential target genes were regulated by the three miRNAs, and 291 of the potential genes were involved in a protein interaction network. Gene ontology annotation indicated that 291 potential genes were enriched in 14 biological process annotations and 2 molecular function annotations. miRNA dysregulation may be involved in early childhood obesity.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , MicroRNAs/genetics , Pediatric Obesity/genetics , Child , Child, Preschool , Computational Biology , Gene Ontology , Humans , MicroRNAs/metabolism , Protein Interaction Maps
13.
Int J Mol Sci ; 18(7)2017 Jul 14.
Article in English | MEDLINE | ID: mdl-28708070

ABSTRACT

MicroRNAs (miRNAs) are important post-transcriptional regulators during adipocyte adipogenesis. MiR-206-3p, a tissue-specific miRNA, is absent in white adipocytes. In this study, we examined the roles of mmu-miR-206-3p in the adipogenic differentiation of 3T3-L1 preadipocytes. The miR-206-3p expression has shown an apparent decreasing trend after induction, and sustained low expression throughout the differentiation of 3T3-L1 cells. miR-206-3p blocked the adipogenic differentiation of 3T3-L1 cells by attenuating c-Met expression; the inhibition effect of miR-206 to the adipogenic differentiation can be counteracted by restoring c-Met expression. In addition, miR-206-3p decreased the phosphorylation of Akt, which is the downstream effector of c-Met in the PI3K/Akt signaling pathway. These data indicate that miR-206-3p inhibits adipocyte adipogenesis through silencing c-Met and subsequently inactivating the PI3K/Akt signaling pathway.


Subject(s)
Adipogenesis/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , 3T3-L1 Cells , Animals , Base Sequence , Down-Regulation/genetics , Mice , MicroRNAs/genetics
14.
Cell Physiol Biochem ; 42(1): 416-426, 2017.
Article in English | MEDLINE | ID: mdl-28558369

ABSTRACT

BACKGROUND/AIMS: Obesity is a major contributor to the growing prevalence of metabolic and cardiovascular diseases. This study was designed to investigate the effect of folic acid (FA) on obese mice by detecting the genome-wide expression profile of lncRNAs and mRNAs in the heart. METHODS: Heart samples were collected from mice fed with standard diet (SD), high-fat diet (HFD) and high-fat diet with FA intake (HFDF). LncRNAs and mRNAs between HFD and HFDF group were analyzed by lncRNA microarray. Nine lncRNAs and mRNAs were validated using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Bioinformatics prediction was used to investigate the potential function of these differentially expressed lncRNAs. Co-expresson analysis was used to determine the transcriptional regulatory relationship of differentially expressed lncRNAs and mRNAs between two groups. RESULTS: The expression of 58,952 lncRNAs and 20,145 mRNAs in HFD and HFDF groups was profiled by using microarrays. Gene Ontology and pathway analyses indicated that the biological functions of differentially expressed mRNAs were related to inflammation, energy metabolism, and cell differentiation. Co-expression networks composed of lncRNAs and mRNAs were also constructed to investigate the potential regulatory roles of differentially expressed lncRNAs on mRNAs. LncRNAs, namely, NONMMUT033847, NONMMUT070811, and NONMMUT015327, were validated through qRT-PCR, and these lncRNAs may be important factors regulating inflammation, energy metabolism, and cell differentiation. The expression levels of Dnajb1, Egr2, Hba-a1, Il1ß, Cxcl2, and Tnfsf9 were significantly different between HFD and HFDF. CONCLUSIONS: Results suggested that FA may improve the cardiovascular function of obesity and contribute to those lncRNAs associated with inflammation and cell differentiation. In a nutshell, the present study identified a panel of lncRNAs and mRNAs that may be potential biomarkers or drug targets relevant to the high-fat diet related obesity.


Subject(s)
Folic Acid/pharmacology , RNA, Long Noncoding/metabolism , Transcriptome/drug effects , Adipokines/blood , Animals , Body Weight , Cell Differentiation/genetics , Diet, High-Fat , Dietary Supplements , Energy Metabolism/genetics , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
15.
Mol Neurobiol ; 54(8): 6304-6316, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27714636

ABSTRACT

Dishevelled (DVL/Dvl) genes play roles in canonical and noncanonical Wnt signaling, both of which are essential in neural tube closing and are involved in balancing neural progenitor growth and differentiation, or neuroepithelial cell polarity, respectively. In mouse Dvl haploinsufficiency leads to neural tube defects (NTDs), which represent the second most common birth defects. However, DVL genes' genetic contributions in human NTDs are modest. We sought to explore the molecular impact on such genes in human NTDs in a Han Chinese cohort. In 47 cases with NTDs and 61 matched controls, in brain tissues, the DVL1/2 mRNA levels were correlated with the levels of a serine/threonine protein kinase MARK2, and in 20 cases with lumbosacral spina bifida, the mRNA levels of DVL1 and MARK2 were significantly decreased; by contrast, only an intronic rare variant was found. Moreover, in an extended population, we found merely three novel rare missense variants in 1 % of individuals with NTDs. In cell-based assays, Mark2 depletion indeed reduces Dvl gene expression and interrupts neural stem cell (NSCs) growth and differentiation, which are likely to be mediated through a decrease in class IIa HDAC phosphorylation and reduced H3K4ac and H3K27ac occupancies at the Dvl1/2 promoters. Finally, the detections of folate concentration in human brain tissue and NSCs and MEF cells indicates that folate deficiency contributes to the observed decreases in Mark2 and Dvl1 expression. Our present study raises a potential common pathogenicity mechanism in human lumbosacral spina bifida about DVL genes rather than their genetic pathogenic role.


Subject(s)
Dishevelled Proteins/genetics , Histones/metabolism , Protein Serine-Threonine Kinases/genetics , Spinal Dysraphism/genetics , Transcription, Genetic , Acetylation , Cell Proliferation/physiology , Child , Child, Preschool , Dishevelled Proteins/metabolism , Female , Humans , Infant , Infant, Newborn , Male , Neural Stem Cells/metabolism , Neurogenesis/physiology , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Spinal Dysraphism/metabolism
16.
Placenta ; 48: 80-86, 2016 12.
Article in English | MEDLINE | ID: mdl-27871477

ABSTRACT

INTRODUCTION: Neural-tube defects (NTDs) are common birth defects of complex etiology. Although many studies have confirmed a genetic component, the exact mechanism between DNA methylation and NTDs remains unclear. METHODS: In this work, we investigated the alteration of methylation from placental tissues obtained from 152 normal infants or with NTDs in 130 children with neural-tube defects. Genome-wide changes in DNA methylation were measured using the NimbleGen microarray. The expression levels of 12 genes were also determined, and two genes (AKT2 and CDC25C) showed low expression in NTDs by quantitative real-time PCR analysis. Then, the methyhlated region of AKT2 promoter sequences were confirmed by massARRAY. RESULTS: A total of 150 differentially methylated regions (81 low methylated regions and 69 high methylated regions) were selected by microarray. The expression levels of AKT2 and CDC25C showed lower expression in NTDs. And the percentage of methyhlated region of AKT2 promoter were increased in NTDs. CONCLUSIONS: DNA mythelation was one of the possible epigenetic variations correlated with the occurrence of NTDs, and AKT2 may be a candidate gene for NTDs.


Subject(s)
DNA Methylation , Neural Tube Defects/genetics , Placenta/metabolism , Proto-Oncogene Proteins c-akt/genetics , Adolescent , Adult , Female , Humans , Male , Neural Tube Defects/metabolism , Pregnancy , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Young Adult , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
17.
Endocr J ; 63(6): 581-7, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27040725

ABSTRACT

This study aimed to characterize the inflammatory cytokine profiles and further validate the significantly different cytokines in the serum obtained from obese children aged 36-48 months. Four obese children and four lean controls were randomly selected for inflammatory cytokine array assay, in which two cytokines [soluble tumor necrosis factor-α receptors (sTNFRs) 1 and 2] were found to be significantly different. Both cytokines (sTNFR1 and sTNFR2) were then further validated through enzyme-linked immunosorbent assay (ELISA) in 61 obese children and 52 lean children. ELISA results revealed that serum sTNFR1 level in obese children significantly increased (p = 0.003), whereas sTNFR2 did not change significantly (p = 0.069). Stratified analysis by gender showed that only obese girls presented increased sTNFR1 (p = 0.005) and sTNFR2 (p = 0.049) levels. We conclude that serum sTNFR1 is elevated in early childhood obesity. Moreover, serum sTNFR1 and sTNFR2 are associated with obese girls but not obese boys, thereby suggesting that serum sTNFRs in early childhood obesity may be sex related.


Subject(s)
Biomarkers/blood , Pediatric Obesity/blood , Receptors, Tumor Necrosis Factor/blood , Body Mass Index , Child, Preschool , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Inflammation Mediators/blood , Male , Sex Characteristics
18.
J Int Med Res ; 44(1): 99-108, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26786758

ABSTRACT

OBJECTIVE: To describe the deletion patterns and distribution characteristics of the dystrophin gene in a Chinese population of patients with Duchenne muscular dystrophy (DMD) or Becker muscular dystrophy (BMD). METHODS: Patients with DMD/BMD were recruited. Deletions in 19 exons of the dystrophin gene were evaluated using accurate multiplex polymerase chain reaction (PCR). RESULT: Multiplex PCR identified deletions in 238/401 (59.4%) patients with DMD/BMD. Of these, 196 (82.4%) were in the distal hotspot, 32 (13.4%) were in the proximal hotspot, five (2.1%) were in both regions and five (2.1%) were in neither hotspot. Deletions were classified into 54 patterns. Exon 49 was the most frequently deleted. The reading frame rule was upheld for 91.9% of cases. CONCLUSION: Accurate multiplex PCR for 19 exons is an effective diagnostic tool.


Subject(s)
Asian People/genetics , Dystrophin/genetics , Gene Deletion , Adolescent , Child , Child, Preschool , Exons/genetics , Humans , Infant , Male , Muscular Dystrophy, Duchenne/genetics , Open Reading Frames/genetics , Sequence Deletion/genetics
19.
Int J Clin Exp Med ; 8(8): 11930-5, 2015.
Article in English | MEDLINE | ID: mdl-26550106

ABSTRACT

Pulmonary hypertension is characterized by extensive vascular remodelling, leading to increased pulmonary vascular resistance and eventual death due to right heart failure. The pathogenesis of pulmonary hypertension involves vascular endothelial dysfunction and disordered vascular smooth muscle cell (VSMC) proliferation and migration, but the exact processes remain unknown. Sphingosine 1-phosphate (S1P) is a bioactive lysophospholipid involved in a wide spectrum of biological processes. S1P has been shown to regulate VSMC proliferation and migration and vascular tension via a family of five S1P G-protein-coupled receptors (S1P1-SIP5). S1P has been shown to have both a vasoconstrictive and vasodilating effect. The S1P receptors S1P1 and S1P3 promote, while S1P2 inhibits VSMC proliferation and migration in vitro in response to S1P. Moreover, it has been reported recently that sphingosine kinase 1 and S1P2 inhibitors might be useful therapeutic agents in the treatment of empirical pulmonary hypertension. The sphingosine kinase 1/S1P signalling pathways may play a role in the pathogenesis of pulmonary hypertension. Modulation of this pathway may offer novel therapeutic strategies.

20.
Endocrine ; 47(3): 749-57, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25027295

ABSTRACT

Insulin resistance (IR) is a major risk for the development of type 2 diabetes (T2D). Numerous studies have been conducted to determine the relationship of ADIPOQ G276T with IR and blood glucose, but the results are conflicting. We conducted a meta-analysis in this work to further investigate such an association. Published works were retrieved from PubMed and Embase databases. A pooled standardized mean difference (SMD) with 95 % confidence interval (CI) was calculated under a dominant model using a random effect model. Twenty-five studies involving 7,480 subjects were included in the meta-analysis. A significant association of G276T polymorphism with homeostasis model assessment insulin resistance index (HOMA-IR) was observed after excluding studies with obvious heterogeneity by performing influence analysis (SMD = -0. 08, 95 % CI -0.15 to -0.01, p = 0.019) and Galbraith plot analysis (SMD = -0.08, 95 % CI -0.14 to -0.02, p = 0.005). In the subgroup analysis, Asians with TG/TT genotypes were found to have a significantly decreased degree of HOMA-IR (SMD = -0.13, 95 % CI -0.20 to -0.07, p < 0.001). The association of G276T with IR or blood glucose for the subgroups (obese, male, and T2D) was also significant. However, the results may not be reliable for small sample sizes. The current meta-analysis suggested that the G276T polymorphism may genetically affect IR using HOMA-IR as an index, and the effect of this polymorphism on IR particularly exists in Asians.


Subject(s)
Adiponectin/genetics , Blood Glucose/genetics , Polymorphism, Single Nucleotide , Genetic Association Studies , Genotype , Humans , Insulin Resistance/genetics
SELECTION OF CITATIONS
SEARCH DETAIL