TGFβ1-Induced Baf60c Regulates both Smooth Muscle Cell Commitment and Quiescence

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. TGFβ1 alone, or with PDGF-BB, induces differentiation of rMAPCs to SMCs, which expressed structural SMC proteins, including α-smooth muscle actin (αSMA), 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 TGFβ1 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. TGFβ1 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 TGFβ1, 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 TGFβ-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.

[1]  F. Prósper,et al.  MAPC culture conditions support the derivation of cells with nascent hypoblast features from bone marrow and blastocysts. , 2012, Journal of molecular cell biology.

[2]  Zhenguo Wu,et al.  Signal‐dependent incorporation of MyoD–BAF60c into Brg1‐based SWI/SNF chromatin‐remodelling complex , 2012, The EMBO journal.

[3]  M. Trotter,et al.  Generation of human vascular smooth muscle subtypes provides insight into embryological origin-dependent disease susceptibility , 2012, Nature Biotechnology.

[4]  A. Luttun,et al.  Differentiation Potential of Human Postnatal Mesenchymal Stem Cells, Mesoangioblasts, and Multipotent Adult Progenitor Cells Reflected in Their Transcriptome and Partially Influenced by the Culture Conditions , 2011, Stem cells.

[5]  J. M. Rodenberg,et al.  The SWI/SNF Chromatin Remodeling Complex Regulates Myocardin-Induced Smooth Muscle–Specific Gene Expression , 2009, Arteriosclerosis, thrombosis, and vascular biology.

[6]  Benoit G. Bruneau,et al.  Directed transdifferentiation of mouse mesoderm to heart tissue by defined factors , 2009, Nature.

[7]  B. Amendt,et al.  Pitx2 is functionally important in the early stages of vascular smooth muscle cell differentiation , 2008, The Journal of cell biology.

[8]  M. Westerfield,et al.  Smarcd3 Regulates the Timing of Zebrafish Myogenesis Onset* , 2008, Journal of Biological Chemistry.

[9]  A. Luttun,et al.  Comparative transcriptome analysis of embryonic and adult stem cells with extended and limited differentiation capacity , 2007, Genome Biology.

[10]  A. Luttun,et al.  In vitro and in vivo arterial differentiation of human multipotent adult progenitor cells , 2006 .

[11]  Claudio Cobelli,et al.  Significance analysis of microarray transcript levels in time series experiments , 2007, BMC Bioinformatics.

[12]  Joan M. Taylor,et al.  Smooth muscle cell-specific transcription is regulated by nuclear localization of the myocardin-related transcription factors. , 2007, American journal of physiology. Heart and circulatory physiology.

[13]  J. Rossant,et al.  Baf60c is a nuclear Notch signaling component required for the establishment of left–right asymmetry , 2007, Proceedings of the National Academy of Sciences.

[14]  Robert T Tranquillo,et al.  Cytokine-induced differentiation of multipotent adult progenitor cells into functional smooth muscle cells. , 2006, The Journal of clinical investigation.

[15]  M. Jinnin,et al.  Characterization of SIS3, a Novel Specific Inhibitor of Smad3, and Its Effect on Transforming Growth Factor-β1-Induced Extracellular Matrix Expression , 2006, Molecular Pharmacology.

[16]  Jeffrey L. Wrana,et al.  Baf60c is essential for function of BAF chromatin remodelling complexes in heart development , 2004, Nature.

[17]  Patrick J. Paddison,et al.  Cloning of short hairpin RNAs for gene knockdown in mammalian cells , 2004, Nature Methods.

[18]  G. Owens,et al.  Molecular regulation of vascular smooth muscle cell differentiation in development and disease. , 2004, Physiological reviews.

[19]  Markella Ponticos,et al.  Regulation of Collagen Type I in Vascular Smooth Muscle Cells by Competition between Nkx2.5 and δEF1/ZEB1 , 2004, Molecular and Cellular Biology.

[20]  C. Simone,et al.  p38 pathway targets SWI-SNF chromatin-remodeling complex to muscle-specific loci , 2004, Nature Genetics.

[21]  P. Ward,et al.  Conservation of Sox4 gene structure and expression during chicken embryogenesis. , 2003, Gene.

[22]  J. Miano,et al.  Serum response factor: toggling between disparate programs of gene expression. , 2003, Journal of molecular and cellular cardiology.

[23]  Da-Zhi Wang,et al.  Myocardin Is a Key Regulator of CArG-Dependent Transcription of Multiple Smooth Muscle Marker Genes , 2003, Circulation research.

[24]  M. Yaniv,et al.  When the SWI/SNF complex remodels … the cell cycle , 2001, Oncogene.

[25]  G. Owens,et al.  CArG elements control smooth muscle subtype-specific expression of smooth muscle myosin in vivo. , 2001, The Journal of clinical investigation.

[26]  Dean Y. Li,et al.  Arteriovenous malformations in mice lacking activin receptor-like kinase-1 , 2000, Nature Genetics.

[27]  A. Porteu,et al.  An overlapping CArG/octamer element is required for regulation of desmin gene transcription in arterial smooth muscle cells. , 2000, Developmental biology.

[28]  J. Martin,et al.  The contribution of airway smooth muscle to airway narrowing and airway hyperresponsiveness in disease. , 2000, The European respiratory journal.

[29]  B. Hogan,et al.  Murine forkhead/winged helix genes Foxc1 (Mf1) and Foxc2 (Mfh1) are required for the early organogenesis of the kidney and urinary tract. , 2000, Development.

[30]  P. Donahoe,et al.  Activin receptor-like kinase 1 modulates transforming growth factor-beta 1 signaling in the regulation of angiogenesis. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[31]  B. Hogan,et al.  Roles for the winged helix transcription factors MF1 and MFH1 in cardiovascular development revealed by nonallelic noncomplementation of null alleles. , 1999, Developmental biology.

[32]  C. Betsholtz,et al.  Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. , 1999, Development.

[33]  B. Brooke,et al.  Defective angiogenesis in mice lacking endoglin. , 1999, Science.

[34]  G. Owens,et al.  Regulation of smooth muscle alpha-actin expression in vivo is dependent on CArG elements within the 5' and first intron promoter regions. , 1999, Circulation research.

[35]  C. Deng,et al.  Angiogenesis defects and mesenchymal apoptosis in mice lacking SMAD5. , 1999, Development.

[36]  C. Little,et al.  Morphogenesis of the First Blood Vessels , 1998, Annals of the New York Academy of Sciences.

[37]  C. Heldin,et al.  Identification and Functional Characterization of a Smad Binding Element (SBE) in the JunB Promoter That Acts as a Transforming Growth Factor-β, Activin, and Bone Morphogenetic Protein-inducible Enhancer* , 1998, The Journal of Biological Chemistry.

[38]  Denis Vivien,et al.  Direct binding of Smad3 and Smad4 to critical TGFβ‐inducible elements in the promoter of human plasminogen activator inhibitor‐type 1 gene , 1998, The EMBO journal.

[39]  M. DeRuiter,et al.  Neural crest cell contribution to the developing circulatory system: implications for vascular morphology? , 1998, Circulation research.

[40]  R R Markwald,et al.  Embryonic endothelial cells transdifferentiate into mesenchymal cells expressing smooth muscle actins in vivo and in vitro. , 1997, Circulation research.

[41]  E. Olson,et al.  Expression of the SM22alpha promoter in transgenic mice provides evidence for distinct transcriptional regulatory programs in vascular and visceral smooth muscle cells , 1996, The Journal of cell biology.

[42]  J. Osbourn,et al.  Cloning and analysis of the promoter region of the rat SM22 alpha gene. , 1995, The Biochemical journal.

[43]  J. Osbourn,et al.  Cloning and analysis of the promoter region of the rat SM22α gene , 1995 .

[44]  A. Kulkarni,et al.  Defective haematopoiesis and vasculogenesis in transforming growth factor-beta 1 knock out mice. , 1995, Development.

[45]  D. Foster,et al.  The 5'-flanking region of the mouse vascular smooth muscle alpha-actin gene contains evolutionarily conserved sequence motifs within a functional promoter. , 1990, The Journal of biological chemistry.

[46]  R. Ross,et al.  A platelet-dependent serum factor that stimulates the proliferation of arterial smooth muscle cells in vitro. , 1974, Proceedings of the National Academy of Sciences of the United States of America.

[47]  C. Verfaillie,et al.  Isolation procedure and characterization of multipotent adult progenitor cells from rat bone marrow. , 2010, Methods in molecular biology.

[48]  G. Owens,et al.  Transforming growth factor-beta1 signaling contributes to development of smooth muscle cells from embryonic stem cells. , 2004, American journal of physiology. Cell physiology.

[49]  R. Ross,et al.  Human atherosclerosis. I. Cell constitution and characteristics of advanced lesions of the superficial femoral artery. , 1984, The American journal of pathology.

[50]  R. Ross,et al.  The smooth muscle cell in culture. , 1979, Physiological reviews.

[51]  E. Morrisey,et al.  Cells Gata-6 Regulates Genes Promoting Synthetic Functions in Vascular Smooth Muscle Gata-6 Regulates Genes Promoting Synthetic Functions in Vascular Smooth Muscle Cells Methods Generation and Characterization of Replication-defective Recombinant Adenoviruses Microarray Analysis of Genes Regulated b , 2022 .

[52]  G. Owens,et al.  Transforming growth factor- (cid:1) 1 signaling contributes to development of smooth muscle cells from embryonic stem cells , 2022 .