2018
DOI: 10.1007/s12035-018-1444-7
|View full text |Cite
|
Sign up to set email alerts
|

FOXG1 Regulates PRKAR2B Transcriptionally and Posttranscriptionally via miR200 in the Adult Hippocampus

Abstract: Rett syndrome is a complex neurodevelopmental disorder that is mainly caused by mutations in MECP2 . However, mutations in FOXG1 cause a less frequent form of atypical Rett syndrome, called FOXG1 syndrome. FOXG1 is a key transcription factor crucial for forebrain development, where it maintains the balance between progenitor proliferation and neuronal differentiation. Using genome-wide small RNA sequencing and quantitative proteomics, we identified that FOXG1 affec… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

4
18
1

Year Published

2019
2019
2023
2023

Publication Types

Select...
8
1

Relationship

2
7

Authors

Journals

citations
Cited by 20 publications
(23 citation statements)
references
References 65 publications
4
18
1
Order By: Relevance
“…We also confirm the nuclear localization of DDX5 and DDX17 in both PSCs and their neural derivatives. This nuclear localization profile is also consistent with previous reports on DDX5 and DDX17 (Lamm, Nicol et al 1996, Kahlina, Goren et al 2004, Alqahtani, Gopal et al 2016, Weise, Arumugam et al 2019, whereas other RNA helicases such as DDX3 (Sekiguchi, Iida et al 2004, Vakilian, Mirzaei et al 2015 and DDX6 (Smillie andSommerville 2002, Kami, Kitani et al 2018) have been identified to mostly localize in the cytoplasm. It is yet to be determined whether DDX5 and DDX17 can possess functions in the nucleus apart from being transcriptional coregulators (Dardenne, Polay Espinoza et al 2014) and microRNA processors (Hong, Noh et al 2013, Kao, Cheng et al 2019, Weise, Arumugam et al 2019.…”
Section: Discussionsupporting
confidence: 92%
“…We also confirm the nuclear localization of DDX5 and DDX17 in both PSCs and their neural derivatives. This nuclear localization profile is also consistent with previous reports on DDX5 and DDX17 (Lamm, Nicol et al 1996, Kahlina, Goren et al 2004, Alqahtani, Gopal et al 2016, Weise, Arumugam et al 2019, whereas other RNA helicases such as DDX3 (Sekiguchi, Iida et al 2004, Vakilian, Mirzaei et al 2015 and DDX6 (Smillie andSommerville 2002, Kami, Kitani et al 2018) have been identified to mostly localize in the cytoplasm. It is yet to be determined whether DDX5 and DDX17 can possess functions in the nucleus apart from being transcriptional coregulators (Dardenne, Polay Espinoza et al 2014) and microRNA processors (Hong, Noh et al 2013, Kao, Cheng et al 2019, Weise, Arumugam et al 2019.…”
Section: Discussionsupporting
confidence: 92%
“…Furthermore, Foxg1 itself has been reported to be one of the miR-200 targets in other model systems (Choi et al, 2008;Garaffo et al, 2015;Zeng et al, 2016), suggesting that FOXG1 expression levels may also be regulated through a miR-200-dependent feedback loop. RNA-seq following miR-200 overexpression and intersection of this cohort with RNA-seq datasets of the Foxg1-heterozygous hippocampus RNA-seq revealed an overlap of 35 potential target genes (Weise et al, 2019). In particular, cAMP-dependent Protein Kinase Type II-Beta Regulatory Subunit (Prkar2b) was identified as a common target for miR-200 and FOXG1 in N2a and hippocampal cells.…”
Section: Post-transcriptional Regulation Via Mirna Processing Pathwaysmentioning
confidence: 93%
“…Further evidence that FOXG1 does not exclusively act at the chromatin comes from an interactome study which demonstrated that the fraction of FOXG1-interacting proteins in N2a cells (Weise et al, 2019) was enriched for proteins affecting post-transcriptional regulation. For example, FOXG1 associates with the microprocessor complex via interaction with DEAD Box Polypeptide 5 (DDX5), demonstrating the role of the FOXG1/DDX5 axis in miRNA biogenesis.…”
Section: Post-transcriptional Regulation Via Mirna Processing Pathwaysmentioning
confidence: 99%
“…Additional development and cell-type specific co-factors regulate microprocessor activity. In addition to MeCP2, a major atypical RTT-pathogenic protein, FOXG1, is recruited to Drosha to influence miRNA biogenesis, implicating the importance of this process to RTT pathology ( Weise et al, 2019 ).…”
Section: Subsectionsmentioning
confidence: 99%