Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 2448: 217-234, 2022.
Article in English | MEDLINE | ID: mdl-35167100

ABSTRACT

Brown adipose tissue (BAT) is a thermoregulatory fat with energy-consuming properties. The location and heterogeneity of this tissue makes it complicated to sample before and after interventions in humans, and an in vitro model for mechanistic and molecular studies is therefore of great value. We here describe a protocol for isolation of progenitors from the stromal vascular fraction of BAT biopsies obtained surgically from adult humans. We further present how these cells are differentiated in vitro and finally how they are characterized for thermogenic capacity. Methods for characterization described here include norepinephrine-induced thermogenic gene expression using qPCR; norepinephrine-induced mitochondrial uncoupling using the Seahorse XFe96 Analyzer, and norepinephrine-induced expression of UCP1 using the RNAscope® Technology.


Subject(s)
Adipocytes, Brown , Mitochondrial Proteins , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Cell Differentiation , Humans , Mitochondrial Proteins/metabolism , Thermogenesis , Uncoupling Protein 1/genetics
2.
Nat Commun ; 12(1): 2431, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893273

ABSTRACT

Insulin resistance and lower muscle quality (strength divided by mass) are hallmarks of type 2 diabetes (T2D). Here, we explore whether alterations in muscle stem cells (myoblasts) from individuals with T2D contribute to these phenotypes. We identify VPS39 as an important regulator of myoblast differentiation and muscle glucose uptake, and VPS39 is downregulated in myoblasts and myotubes from individuals with T2D. We discover a pathway connecting VPS39-deficiency in human myoblasts to impaired autophagy, abnormal epigenetic reprogramming, dysregulation of myogenic regulators, and perturbed differentiation. VPS39 knockdown in human myoblasts has profound effects on autophagic flux, insulin signaling, epigenetic enzymes, DNA methylation and expression of myogenic regulators, and gene sets related to the cell cycle, muscle structure and apoptosis. These data mimic what is observed in myoblasts from individuals with T2D. Furthermore, the muscle of Vps39+/- mice display reduced glucose uptake and altered expression of genes regulating autophagy, epigenetic programming, and myogenesis. Overall, VPS39-deficiency contributes to impaired muscle differentiation and reduced glucose uptake. VPS39 thereby offers a therapeutic target for T2D.


Subject(s)
Autophagy-Related Proteins/genetics , Autophagy/genetics , Cell Differentiation/genetics , Diabetes Mellitus, Type 2/genetics , Epigenomics/methods , Myoblasts/metabolism , Stem Cells/metabolism , Vesicular Transport Proteins/genetics , Animals , Autophagy-Related Proteins/deficiency , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Epigenesis, Genetic/genetics , Female , Gene Expression Profiling/methods , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle Development/genetics , Vesicular Transport Proteins/deficiency
3.
Stem Cell Res Ther ; 10(1): 26, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30646953

ABSTRACT

BACKGROUND: Sex differences are known to impact muscle phenotypes, metabolism, and disease risk. Skeletal muscle stem cells (satellite cells) are important for muscle repair and to maintain functional skeletal muscle. Here we studied, for the first time, effects of sex on DNA methylation and gene expression in primary human myoblasts (activated satellite cells) before and after differentiation into myotubes. METHOD: We used an array-based approach to analyse genome-wide DNA methylation and gene expression in myoblasts and myotubes from 13 women and 13 men. The results were followed up with a reporter gene assay. RESULTS: Genome-wide DNA methylation and gene expression differences between the sexes were detected in both myoblasts and myotubes, on the autosomes as well as the X-chromosome, despite lack of exposure to sex hormones and other factors that differ between sexes. Pathway analysis revealed higher expression of oxidative phosphorylation and other metabolic pathways in myoblasts from women compared to men. Oxidative phosphorylation was also enriched among genes with higher expression in myotubes from women. Forty genes in myoblasts and 9 in myotubes had differences in both DNA methylation and gene expression between the sexes, including LAMP2 and SIRT1 in myoblasts and KDM6A in myotubes. Furthermore, increased DNA methylation of LAMP2 promoter had negative effects on reporter gene expression. Five genes (CREB5, RPS4X, SYAP1, XIST, and ZRSR2) showed differential DNA methylation and gene expression between the sexes in both myoblasts and myotubes. Interestingly, differences in DNA methylation and expression between women and men were also found during differentiation (myoblasts versus myotubes), e.g., in genes involved in energy metabolism. Interestingly, more DNA methylation changes occur in women compared to men on autosomes. CONCLUSION: All together, we show that epigenetic and transcriptional differences exist in human myoblasts and myotubes as well as during differentiation between women and men. We believe that these intrinsic differences might contribute to sex dependent differences in muscular phenotypes.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation, Developmental/genetics , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Humans , Middle Aged , Sex Factors
4.
Genome Med ; 9(1): 47, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28545587

ABSTRACT

BACKGROUND: Skeletal muscle is one of the primary tissues involved in the development of type 2 diabetes (T2D). The close association between obesity and T2D makes it difficult to isolate specific effects attributed to the disease alone. Therefore, here we set out to identify and characterize intrinsic properties of myocytes, associated independently with T2D or obesity. METHODS: We generated and analyzed RNA-seq data from primary differentiated myotubes from 24 human subjects, using a factorial design (healthy/T2D and non-obese/obese), to determine the influence of each specific factor on genome-wide transcription. This setup enabled us to identify intrinsic properties, originating from muscle precursor cells and retained in the corresponding myocytes. Bioinformatic and statistical methods, including differential expression analysis, gene-set analysis, and metabolic network analysis, were used to characterize the different myocytes. RESULTS: We found that the transcriptional program associated with obesity alone was strikingly similar to that induced specifically by T2D. We identified a candidate epigenetic mechanism, H3K27me3 histone methylation, mediating these transcriptional signatures. T2D and obesity were independently associated with dysregulated myogenesis, down-regulated muscle function, and up-regulation of inflammation and extracellular matrix components. Metabolic network analysis identified that in T2D but not obesity a specific metabolite subnetwork involved in sphingolipid metabolism was transcriptionally regulated. CONCLUSIONS: Our findings identify inherent characteristics in myocytes, as a memory of the in vivo phenotype, without the influence from a diabetic or obese extracellular environment, highlighting their importance in the development of T2D.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Histones/metabolism , Muscle Fibers, Skeletal/physiology , Obesity/genetics , Adult , Computational Biology , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Inflammation , Male , Methylation , Middle Aged , Muscle Development , Muscle Fibers, Skeletal/pathology , Obesity/pathology , Obesity/physiopathology , Sequence Analysis, RNA , Sphingolipids/metabolism
5.
PLoS One ; 8(6): e66628, 2013.
Article in English | MEDLINE | ID: mdl-23805253

ABSTRACT

Both aging and physical inactivity are associated with increased development of insulin resistance whereas physical activity has been shown to promote increased insulin sensitivity. Here we investigated the effects of physical activity level on aging-associated insulin resistance in myotubes derived from human skeletal muscle satellite cells. Satellite cells were obtained from young (22 yrs) normally active or middle-aged (56.6 yrs) individuals who were either lifelong sedentary or lifelong active. Both middle-aged sedentary and middle-aged active myotubes had increased p21 and myosin heavy chain protein expression. Interestingly MHCIIa was increased only in myotubes from middle-aged active individuals. Middle-aged sedentary cells had intact insulin-stimulated Akt phosphorylation however, the same cell showed ablated insulin-stimulated glucose uptake and GLUT4 translocation to the plasma membrane. On the other hand, middle-aged active cells retained both insulin-stimulated increases in glucose uptake and GLUT4 translocation to the plasma membrane. Middle-aged active cells also had significantly higher mRNA expression of GLUT1 and GLUT4 compared to middle-aged sedentary cells, and significantly higher GLUT4 protein. It is likely that physical activity induces a number of stable adaptations, including increased GLUT4 expression that are retained in cells ex vivo and protect, or delay the onset of middle-aged-associated insulin resistance. Additionally, a sedentary lifestyle has an impact on the metabolism of human myotubes during aging and may contribute to aging-associated insulin resistance through impaired GLUT4 localization.


Subject(s)
Aging , Exercise , Gene Expression Regulation/physiology , Glucose Transporter Type 4/genetics , Insulin Resistance , Satellite Cells, Skeletal Muscle/metabolism , Cells, Cultured , Gene Expression Profiling , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance/genetics , Middle Aged , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA/isolation & purification , RNA/metabolism , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...