2013
DOI: 10.1074/jbc.m113.455899
|View full text |Cite
|
Sign up to set email alerts
|

Epigenetic Switching by the Metabolism-sensing Factors in the Generation of Orexin Neurons from Mouse Embryonic Stem Cells

Abstract: Background: Orexin plays a central role in the integration of sleep/wake states and feeding behaviors. Results: Orexin neurons were induced from pluripotent stem cells by supplementation of ManNAc. Conclusion: ManNAc induced switching of epigenetic factors from Sirt1/Ogt to Mgea5 at Hcrt gene locus. Significance: This study will be useful to investigate molecular mechanism in the orexin system and development of regenerative medicine.

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

2
69
0

Year Published

2015
2015
2021
2021

Publication Types

Select...
6
2
1

Relationship

0
9

Authors

Journals

citations
Cited by 71 publications
(71 citation statements)
references
References 60 publications
2
69
0
Order By: Relevance
“…O-GlcNAcylation modifications appear to influence the determination of specific neuronal identity. During mESC differentiation, OGA co-localizes with the histone acetyltransferase p300 on the neuropeptide encoding gene orexin in orexin neurons, while OGT co-localizes with histone deacetylase sirtuin 1 (SIRT1) in non-orexin expressing cells (65). Due to the direct link between HBP and O-GlcNAcylation, we propose that metabolic fluctuations induced by specific microenvironmental differentiation drivers, such as hypoxia, could participate in establishing distinct epigenome patterns associated with cell fate and function acquisitions and maintenance.…”
Section: O-glcnacylationmentioning
confidence: 99%
See 1 more Smart Citation
“…O-GlcNAcylation modifications appear to influence the determination of specific neuronal identity. During mESC differentiation, OGA co-localizes with the histone acetyltransferase p300 on the neuropeptide encoding gene orexin in orexin neurons, while OGT co-localizes with histone deacetylase sirtuin 1 (SIRT1) in non-orexin expressing cells (65). Due to the direct link between HBP and O-GlcNAcylation, we propose that metabolic fluctuations induced by specific microenvironmental differentiation drivers, such as hypoxia, could participate in establishing distinct epigenome patterns associated with cell fate and function acquisitions and maintenance.…”
Section: O-glcnacylationmentioning
confidence: 99%
“…The hexosamine biosynthesis pathway (HBP) is a nutrient-sensing pathway that depends on glucose, glutamine, and cytosolic Ac-CoA levels (Figure 2 Conversely, a decrease in O-GlcNAcylation disrupts self-renewal and reprogramming (57), but promotes differentiation in neuronal cells (63)(64)(65). Moreover, CPTFs such as OCT4 and SOX2 harbor an O-GlcNAcylation motif that is quickly erased upon differentiation (57).…”
Section: Hexosamine Biosynthesis and Fate Transitionsmentioning
confidence: 99%
“…ManNAc has been shown to successfully generate functional orexin neurons from mouse ESCs, and the key step in the differentiation pathway is an epigenetic switch on histones from a hypoacetylated state with unidentified OGlcNAcylated nuclear proteins to a hyperacetylated state at T-DMRs. ManNAc causes the dislocation of Ogt/Sirt1/Sin3A/ Ezh2 from the T-DMRs of the Hcrt gene locus and the recruitment of p300, CBP, and Mgea5 [108].…”
Section: Cell Fate Determination Of Neuronal Subtypes By Sirt1mentioning
confidence: 99%
“…Intriguingly, OGA is a bifunctional enzyme harboring O-GlcNAc cleavage activity as well as HAT activity, implying an intrinsic relationship between histone O-GlcNAcylation and acetylation. OGA HAT activity has been implicated in orexin neurogenesis [43]. However, we are just beginning to understand the function of histone O-GlcNAcylation as part of the epigenetic code.…”
Section: Protein O-glcnacylation In Epigeneticsmentioning
confidence: 99%