2017
DOI: 10.1073/pnas.1705755114
|View full text |Cite
|
Sign up to set email alerts
|

Smad7 enables STAT3 activation and promotes pluripotency independent of TGF-β signaling

Abstract: Smad7 is a negative feedback product of TGF-β superfamily signaling and fine tunes a plethora of pleiotropic responses induced by TGF-β ligands. However, its noncanonical functions independent of TGF-β signaling remain to be elucidated. Here, we show that Smad7 activates signal transducers and activators of transcription 3 (STAT3) signaling in maintaining mouse embryonic stem cell pluripotency in a manner independent of the TGF-β receptors, yet dependent on the leukemia inhibitory factor (LIF) coreceptor glyco… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

2
41
0

Year Published

2018
2018
2024
2024

Publication Types

Select...
8
1

Relationship

1
8

Authors

Journals

citations
Cited by 52 publications
(43 citation statements)
references
References 54 publications
(48 reference statements)
2
41
0
Order By: Relevance
“…Considering REGγ knockout mice can survive and breed normally, we assume that targeting REGγ may be an alternative option for ATC therapy in the future with less side effects than inhibition of the conventional ubiquitin-proteasome degradation system. In addition, Smad7 has TGF-β-independent function in promoting pluripotency by amplifying STAT3 activation [34]. Thus, our study suggests: (1) targeting Smad7 downstream of TGF-β but not directly affecting receptor levels, may be an economical approach.…”
Section: Discussionmentioning
confidence: 80%
“…Considering REGγ knockout mice can survive and breed normally, we assume that targeting REGγ may be an alternative option for ATC therapy in the future with less side effects than inhibition of the conventional ubiquitin-proteasome degradation system. In addition, Smad7 has TGF-β-independent function in promoting pluripotency by amplifying STAT3 activation [34]. Thus, our study suggests: (1) targeting Smad7 downstream of TGF-β but not directly affecting receptor levels, may be an economical approach.…”
Section: Discussionmentioning
confidence: 80%
“…Obesity and older age are high-risk factors for developing diabetes [ 1 ], and over-production of TGF-β in the hypothalamic POMC neurons promotes glucose disorders under these conditions [ 8 ]. As an inhibitor of TGF-β signaling [ 16 ], the role of central Smad7 in glucose metabolism has not been investigated, although many functions of Smad7 in peripheral tissues have been reported [ 12 ]. Based on the well-known relationship between TGF-β and Smad7 [ 16 ], we speculated that hypothalamic Smad7 expression levels might be high in response to the over-production of TGF-β in HFD-fed or middle-aged mice.…”
Section: Discussionmentioning
confidence: 99%
“…Once activated, Smad7 binds to TGF-β type 1 receptor (TβR1) and recruits ubiquitin ligase to degrade TβR1 or protein phosphatase 1c (PP1c) to dephosphorylate TβR1 [ 10 , 11 ]. In addition to the TGF-β-dependent regulatory mechanism, Smad7 may also regulate downstream signaling independent of TGF-β [ 12 ]. Smad7 has been shown to affect the regulation of many processes (i.e., via intensifying enteritis, promoting tumor cell growth, and inhibiting hepatic fibrosis) [ [13] , [14] , [15] ].…”
Section: Introductionmentioning
confidence: 99%
“…In early-stage BL, the expression levels of SOX2, OCT4, and NANOG were not signi cantly different according to SOX2 targeting ( Figure 4A). The expression of SOX17 decreased, and the expression of SMAD7, which is known to be involved in ES cell self-renewal and iPSC reprogramming, also decreased (25). KDM8 is directly controlled by SOX2, which is known to regulate embryonic cell proliferation, and its expression is decreased in SOX2-targeted blastocyst ( Figure 3C) (26,27).…”
Section: Effects Of Sox2-targeting Plasmid Microinjection On Embryo Dmentioning
confidence: 98%