Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Sleep ; 37(5): 999-1009, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24790279

ABSTRACT

BACKGROUND: Chronic sleep fragmentation (SF) without sleep curtailment induces increased adiposity. However, it remains unclear whether mobilization, proliferation, and differentiation of adipocyte progenitors (APs) occurs in visceral white adipose tissue (VWAT), and whether nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2) activity plays a role. METHODS: Changes in VWAT depot cell size and AP proliferation were assessed in wild-type and Nox2 null male mice exposed to SF and control sleep (SC). To assess mobilization, proliferation, and differentiation of bone marrow mesenchymal stem cells (BM-MSC), Sca-1+ bone marrow progenitors were isolated from GFP+ or RFP+ mice, and injected intravenously to adult male mice (C57BL/6) previously exposed to SF or SC. RESULTS: In comparison with SC, SF was associated with increased weight accrual at 3 w and thereafter, larger subcutaneous and visceral fat depots, and overall adipocyte size at 8 w. Increased global AP numbers in VWAT along with enhanced AP BrDU labeling in vitro and in vivo emerged in SF. Systemic injections of GFP+ BM-MSC resulted in increased AP in VWAT, as well as in enhanced differentiation into adipocytes in SF-exposed mice. No differences occurred between SF and SC in Nox2 null mice for any of these measurements. CONCLUSIONS: Chronic sleep fragmentation (SF) induces obesity in mice and increased proliferation and differentiation of adipocyte progenitors (AP) in visceral white adipose tissue (VWAT) that are mediated by increased Nox2 activity. In addition, enhanced migration of bone marrow mesenchymal stem cells from the systemic circulation into VWAT, along with AP differentiation, proliferation, and adipocyte formation occur in SF-exposed wild-type but not in oxidase 2 (Nox2) null mice. Thus, Nox2 may provide a therapeutic target to prevent obesity in the context of sleep disorders.


Subject(s)
Adipocytes/cytology , Adipose Tissue, White/cytology , Cell Differentiation , Intra-Abdominal Fat/cytology , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Sleep Deprivation/enzymology , Stem Cells/cytology , Adipocytes/enzymology , Adipose Tissue, White/enzymology , Adiposity , Animals , Cell Movement , Intra-Abdominal Fat/enzymology , Male , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , NADP/metabolism , NADPH Oxidase 2 , NADPH Oxidases/deficiency , Obesity/complications , Obesity/enzymology , Obesity/genetics , Obesity/pathology , Sleep Deprivation/complications , Sleep Deprivation/genetics , Sleep Deprivation/pathology , Stem Cells/enzymology
2.
PLoS One ; 7(5): e37669, 2012.
Article in English | MEDLINE | ID: mdl-22629440

ABSTRACT

Obstructive sleep apnea (OSA) is a prevalent condition and strongly associated with metabolic disorders. Sleep fragmentation (SF) is a major consequence of OSA, but its contribution to OSA-related morbidities is not known. We hypothesized that SF causes specific perturbations in transcriptional networks of visceral fat cells, leading to systemic metabolic disturbances. We simultaneously profiled visceral adipose tissue mRNA and miRNA expression in mice exposed to 6 hours of SF during sleep, and developed a new computational framework based on gene set enrichment and network analyses to merge these data. This approach leverages known gene product interactions and biologic pathways to interrogate large-scale gene expression profiling data. We found that SF induced the activation of several distinct pathways, including those involved in insulin regulation and diabetes. Our integrative methodology identified putative controllers and regulators of the metabolic response during SF. We functionally validated our findings by demonstrating altered glucose and lipid homeostasis in sleep-fragmented mice. This is the first study to link sleep fragmentation with widespread disruptions in visceral adipose tissue transcriptome, and presents a generalizable approach to integrate mRNA-miRNA information for systematic mapping of regulatory networks.


Subject(s)
Adipose Tissue/metabolism , MicroRNAs/genetics , RNA, Messenger/genetics , Sleep Deprivation/genetics , Transcription, Genetic , Animals , Gene Expression Profiling , Homeostasis , Insulin Resistance/physiology , Metabolic Diseases/complications , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Mice , MicroRNAs/metabolism , Obesity/complications , Obesity/genetics , Obesity/metabolism , RNA, Messenger/metabolism , Sleep Deprivation/complications , Sleep Deprivation/metabolism , Transcriptome
3.
J Mol Med (Berl) ; 90(4): 435-45, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22086141

ABSTRACT

Obstructive sleep apnea (OSA) is a prevalent disorder characterized by intermittent hypoxia (IH) during sleep. OSA is strongly associated with obesity and dysregulation of metabolism-yet the molecular pathways linking the effects of IH on adipocyte biology remain unknown. We hypothesized that exposure to IH would activate distinct, time-dependent transcriptional programs in visceral adipose tissue of mice. We exposed 36 mice to IH or normoxia for up to 13 days. We transcriptionally profiled visceral fat tissue harvested from the animals and performed functional enrichment and network analysis on differentially expressed genes. We identified over 3,000 genes with significant expression patterns during the time course of IH exposure. The most enriched pathways mapped to metabolic processes, mitochondrion, and oxidative stress responses. We confirmed the pathophysiological relevance of these findings by demonstrating that mice exposed to chronic IH developed dyslipidemia and underwent significant lipid and protein oxidation within their visceral adipose depots. We applied gene-gene interaction network analysis to identify critical controllers of IH-induced transcriptional programs in adipocytes-these network hubs represent putative targets to modulate the effects of chronic IH on adipose tissue. Our approach to integrate computational methods with gene expression profiling of visceral fat tissue during IH exposure shows promise in helping unravel the mechanistic links between OSA and adipocyte biology.


Subject(s)
Hypoxia/genetics , Hypoxia/metabolism , Intra-Abdominal Fat/metabolism , Transcriptional Activation , Transcriptome , Animals , Dyslipidemias/etiology , Gene Expression Profiling , Gene Regulatory Networks , Hypoxia/complications , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Stress , Sleep Apnea, Obstructive/genetics , Sleep Apnea, Obstructive/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...