Unknown

Dataset Information

0

TGF?1-induced Baf60c regulates both smooth muscle cell commitment and quiescence.


ABSTRACT: Smooth muscle cells (SMCs) play critical roles in a number of diseases; however, the molecular mechanism underlying their development is unclear. Although the role of TGF?1 signaling in SMC development is well established, the downstream molecular signals are not fully understood. We used several rat multipotent adult progenitor cell ((r)MAPC) lines that express levels of Oct4 mRNA similar to hypoblast stem cells (HypoSC), and can differentiate robustly to mesodermal and endodermal cell types. TGF?1 alone, or with PDGF-BB, induces differentiation of rMAPCs to SMCs, which expressed structural SMC proteins, including ?-smooth muscle actin (?SMA), and contribute to the SMC coat of blood vessels in vivo. A genome-wide time-course transcriptome analysis revealed that transcripts of Baf60c, part of the SWI/SNF actin binding chromatin remodeling complex D-3 (SMARCD3/BAF60c), were significantly induced during MAPC-SMC differentiation. We demonstrated that BAF60c is a necessary co-regulator of TGF?1 mediated induction of SMC genes. Knock-down of Baf60c decreased SMC gene expression in rMAPCs whereas ectopic expression of Baf60c was sufficient to commit rMAPCs to SMCs in the absence of exogenous cytokines. TGF?1 activates Baf60c via the direct binding of SMAD2/3 complexes to the Baf60c promoter region. Chromatin- and co-immunoprecipitation studies demonstrated that regulation of SMC genes by BAF60c is mediated via interaction with SRF binding CArG box-containing promoter elements in SMC genes. We noted that compared with TGF?1, Baf60c overexpression in rMAPC yielded SMC with a more immature phenotype. Similarly, Baf60c induced an immature phenotype in rat aortic SMCs marked by increased cell proliferation and decreased contractile marker expression. Thus, Baf60c is important for TGF?-mediated commitment of primitive stem cells (rMAPCs) to SMCs and is associated with induction of a proliferative state of quiescent SMCs. The MAPC-SMC differentiation system may be useful for identification of additional critical (co-)regulators of SMC development.

SUBMITTER: Sohni A 

PROVIDER: S-EPMC3482188 | biostudies-literature | 2012

REPOSITORIES: biostudies-literature

altmetric image

Publications

TGFβ1-induced Baf60c regulates both smooth muscle cell commitment and quiescence.

Sohni Abhishek A   Mulas Francesca F   Ferrazzi Fulvia F   Luttun Aernout A   Bellazzi Riccardo R   Huylebroeck Danny D   Ekker Stephen C SC   Verfaillie Catherine M CM  

PloS one 20121026 10


Smooth muscle cells (SMCs) play critical roles in a number of diseases; however, the molecular mechanism underlying their development is unclear. Although the role of TGFβ1 signaling in SMC development is well established, the downstream molecular signals are not fully understood. We used several rat multipotent adult progenitor cell ((r)MAPC) lines that express levels of Oct4 mRNA similar to hypoblast stem cells (HypoSC), and can differentiate robustly to mesodermal and endodermal cell types. T  ...[more]

Similar Datasets

| S-EPMC4740238 | biostudies-literature
| S-EPMC4931287 | biostudies-other
| S-EPMC4344425 | biostudies-literature
| S-EPMC3984265 | biostudies-literature
| S-EPMC5940262 | biostudies-literature
| S-EPMC4409270 | biostudies-literature
| S-EPMC5025753 | biostudies-literature
| S-EPMC7196185 | biostudies-literature
| S-EPMC5572929 | biostudies-literature
| S-EPMC4231011 | biostudies-literature