Megakaryoblastic Leukemia Factor-1 Transduces Cytoskeletal Signals and Induces Smooth Muscle Cell Differentiation from Undifferentiated Embryonic Stem Cells*
Kevin Du,Mary Chen,Jian Li,John J. Lepore,Patricia Mericko,Michael S. Parmacek
DOI: https://doi.org/10.1074/jbc.m400961200
2004-01-01
Abstract:The SAP domain transcription factor myocardin plays a critical role in the transcriptional program regulating smooth muscle cell differentiation. In this report, we describe the capacity of myocardin to physically associate with megakaryoblastic leukemia factor-1 (MKL1) and characterize the function of MKL1 in smooth muscle cells (SMCs). The MKL1 gene is expressed in most human tissues and myocardin and MKL are co-expressed in SMCs. MKL1 and myocardin physically associate via conserved leucine zipper domains. Overexpression of MKL1 transactivates serum response factor (SRF)-dependent SMC-restricted transcriptional regulatory elements including the SM22α promoter, smooth muscle myosin heavy chain promoter/enhancer, and SM-α-actin promoter/enhancer in non-SMCs. Moreover, forced expression of MKL1 and SRF in undifferentiated SRF-/- embryonic stem cells activates multiple endogenous SMC-restricted genes at levels equivalent to, or exceeding, myocardin. Forced expression of a dominant-negative MKL1 mutant reduces myocardin-induced activation of the SMC-specific SM22α promoter. In NIH3T3 fibroblasts MKL1 localizes to the cytoplasm and translocates to the nucleus in response to serum stimulation, actin treadmilling, and RhoA signaling. In contrast, in SMCs MKL1 is observed exclusively in the nucleus regardless of serum conditions or RhoA signaling. However, when actin polymerization is disrupted MKL1 translocates from the nucleus to the cytoplasm in SMCs. Together, these data were consistent with a model wherein MKL1 transduces signals from the cytoskeleton to the nucleus in SMCs and regulates SRF-dependent SMC differentiation autonomously or in concert with myocardin. The SAP domain transcription factor myocardin plays a critical role in the transcriptional program regulating smooth muscle cell differentiation. In this report, we describe the capacity of myocardin to physically associate with megakaryoblastic leukemia factor-1 (MKL1) and characterize the function of MKL1 in smooth muscle cells (SMCs). The MKL1 gene is expressed in most human tissues and myocardin and MKL are co-expressed in SMCs. MKL1 and myocardin physically associate via conserved leucine zipper domains. Overexpression of MKL1 transactivates serum response factor (SRF)-dependent SMC-restricted transcriptional regulatory elements including the SM22α promoter, smooth muscle myosin heavy chain promoter/enhancer, and SM-α-actin promoter/enhancer in non-SMCs. Moreover, forced expression of MKL1 and SRF in undifferentiated SRF-/- embryonic stem cells activates multiple endogenous SMC-restricted genes at levels equivalent to, or exceeding, myocardin. Forced expression of a dominant-negative MKL1 mutant reduces myocardin-induced activation of the SMC-specific SM22α promoter. In NIH3T3 fibroblasts MKL1 localizes to the cytoplasm and translocates to the nucleus in response to serum stimulation, actin treadmilling, and RhoA signaling. In contrast, in SMCs MKL1 is observed exclusively in the nucleus regardless of serum conditions or RhoA signaling. However, when actin polymerization is disrupted MKL1 translocates from the nucleus to the cytoplasm in SMCs. Together, these data were consistent with a model wherein MKL1 transduces signals from the cytoskeleton to the nucleus in SMCs and regulates SRF-dependent SMC differentiation autonomously or in concert with myocardin. Smooth muscle cells (SMCs) 1The abbreviations used are: SMC, smooth muscle cell; SRF, serum response factor; TCF, ternary complex factor; SM-MHC, smooth muscle-myosin heavy chain; ES, embryonic stem; MRTF, myocardin-related transcription factor; MKL1, megakaryoblastic leukemia factor-1; HA, hemagglutinin; LZ, leucine zipper; CRE, cyclic AMP response element; RT, reverse transcriptase; ChIP, chromatin immunoprecipitation; FL-Myo, FLAG epitope-tagged myocardin; DN, dominant-negative; C(t), cycle threshold; X-Gal, 5-bromo-4-chloro-3-indolyl-β-d-galactopyranoside. are distributed in organ systems and tissues throughout the body where they regulate tonic contractile functions including modulation of vascular tone, bronchial reactivity, gastrointestinal peristalsis, and genitourinary contraction and tone. In contrast to striated muscle cells, SMCs retain the capacity to both proliferate and modulate their phenotype from contractile to synthetic during postnatal development. The plasticity and proliferative capacity of this muscle cell lineage plays a central role in physiological processes including wound healing and angiogenesis. However, modulation of SMC phenotype has also been implicated in the pathogenesis of disease processes including atherosclerosis, restenosis following coronary angioplasty/stent implantation, and asthma. Our group and others have reported that SMC differentiation and modulation of the SMC phenotype is regulated by a serum response factor (SRF)-dependent transcriptional regulatory program (for review see Refs. 1Parmacek M.S. Curr. Top. Dev. Biol. 2001; 51: 69-89Crossref PubMed Google Scholar and 2Miano J.M. J. Mol. Cell. Cardiol. 2003; 35: 577-593Abstract Full Text Full Text PDF PubMed Scopus (488) Google Scholar)). SRF is a MADS box transcription factor that binds to a transcriptional regulatory element designated the CArG box or SRE. The functional properties of SRF and SRF binding sites were initially characterized in promoters regulating growth responsive genes including c-fos and egr-1 (3Treisman R. EMBO J. 1995; 14: 4905-4913Crossref PubMed Scopus (347) Google Scholar, 4Treisman R. Nature. 1995; 376: 468-469Crossref PubMed Scopus (31) Google Scholar, 5Johansen F.E. Prywes R. Biochim. Biophys. Acta. 1995; 1242: 1-10Crossref PubMed Scopus (105) Google Scholar). SRF-mediated activity of growth responsive genes is regulated via post-translational modifications of SRF and ternary complex factors (TCFs) in the ets family that physically associate with both SRF and specific DNA sequences (6Treisman R. Curr. Opin. Genet. Dev. 1994; 4: 96-101Crossref PubMed Scopus (620) Google Scholar). SRF activity is also modulated by intracellular signals including serum, RhoA, and actin treadmilling (7Sotiropoulos A. Gineitis D. Copeland J. Treisman R. Cell. 1999; 98: 159-169Abstract Full Text Full Text PDF PubMed Scopus (574) Google Scholar, 8Treisman R. Alberts A.S. Sahai E. Cold Spring Harbor Symp. Quant. Biol. 1998; 63: 643-651Crossref PubMed Scopus (67) Google Scholar, 9Hill C.S. Wynne J. Treisman R. Cell. 1995; 81: 1159-1170Abstract Full Text PDF PubMed Scopus (1207) Google Scholar, 10Mack C.P. Somlyo A.V. Hautmann M. Somlyo A.P. Owens G.K. J. Biol. Chem. 2001; 276: 341-347Abstract Full Text Full Text PDF PubMed Scopus (333) Google Scholar). Functionally important CArG boxes have been identified in all SMC-restricted transcriptional regulatory elements (1Parmacek M.S. Curr. Top. Dev. Biol. 2001; 51: 69-89Crossref PubMed Google Scholar, 2Miano J.M. J. Mol. Cell. Cardiol. 2003; 35: 577-593Abstract Full Text Full Text PDF PubMed Scopus (488) Google Scholar). Mutation of these CArG boxes abolishes activity of these transcriptional elements. A multimerized copy of the SME-4 CArG box embedded in the SMC-restricted SM22α promoter is sufficient to restrict expression of a reporter gene to arterial SMCs in transgenic mice (11Kim S. Ip H.S. Lu M.M. Clendenin C. Parmacek M.S. Mol. Cell. Biol. 1997; : 2266-2278Crossref PubMed Scopus (190) Google Scholar). Until recently it was not clear how a ubiquitously expressed transcription factor such as SRF could regulate transcription of both growth responsive genes and muscle-restricted differentiation markers. Recent studies revealed that SRF regulates SMC differentiation via direct binding to the recently discovered cardiac- and SMC-restricted SAP (SAF-A/B, Acinus, PIAS) domain transcription factor myocardin (12Chen J. Kitchen C.M. Streb J.W. Miano J.M. J. Mol. Cell. Cardiol. 2002; 34: 1345-1356Abstract Full Text PDF PubMed Google Scholar, 13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar, 14Li S. Wang D.Z. Wang Z. Richardson J.A. Olson E.N. Proc. Natl. Acad. Sci. U. S. A. 2003; 100: 9366-9370Crossref PubMed Scopus (301) Google Scholar, 15Wang Z. Wang D.Z. Pipes G.C. Olson E.N. Proc. Natl. Acad. Sci. U. S. A. 2003; 100: 7129-7134Crossref PubMed Scopus (432) Google Scholar, 16Yoshida T. Sinha S. Dandre F. Wamhoff B.R. Hoofnagle M.H. Kremer B.E. Wang D.Z. Olson E.N. Owens G.K. Circ. Res. 2003; 92: 856-864Crossref PubMed Scopus (310) Google Scholar). Myocardin is a member of an ancient, and diverse, family of SAP domain-containing proteins that have been implicated in high order chromatin organization, apoptotic nuclear condensation and, most recently, RhoA-actin signal transduction (17Miralles F. Posern G. Zaromytidou A.I. Treisman R. Cell. 2003; 113: 329-342Abstract Full Text Full Text PDF PubMed Scopus (1057) Google Scholar, 18Wang D. Chang P.S. Wang Z. Sutherland L. Richardson J.A. Small E. Krieg P.A. Olson E.N. Cell. 2001; 105: 851-862Abstract Full Text Full Text PDF PubMed Scopus (752) Google Scholar, 19Wang D.Z. Li S. Hockemeyer D. Sutherland L. Wang Z. Schratt G. Richardson J.A. Nordheim A. Olson E.N. Proc. Natl. Acad. Sci. U. S. A. 2002; 99: 14855-14860Crossref PubMed Scopus (405) Google Scholar, 20Aravind L. Koonin E.V. Trends Biochem. Sci. 2000; 25: 112-114Abstract Full Text Full Text PDF PubMed Scopus (415) Google Scholar). Forced expression of myocardin activates SMC-restricted transcriptional regulatory elements in non-SMC cells including the SM22α promoter, the smooth muscle myosin heavy chain (SM-MHC) promoter/intragenic enhancer, and the SM-α-actin promoter/intragenic enhancer (12Chen J. Kitchen C.M. Streb J.W. Miano J.M. J. Mol. Cell. Cardiol. 2002; 34: 1345-1356Abstract Full Text PDF PubMed Google Scholar, 13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar, 15Wang Z. Wang D.Z. Pipes G.C. Olson E.N. Proc. Natl. Acad. Sci. U. S. A. 2003; 100: 7129-7134Crossref PubMed Scopus (432) Google Scholar, 16Yoshida T. Sinha S. Dandre F. Wamhoff B.R. Hoofnagle M.H. Kremer B.E. Wang D.Z. Olson E.N. Owens G.K. Circ. Res. 2003; 92: 856-864Crossref PubMed Scopus (310) Google Scholar). Moreover, forced expression of myocardin and SRF in SRF-deficient embryonic stem (ES) cells activates endogenous SMC genes including SM22α, SM-MHC, calponin-h1, and SM-α-actin (13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar). Mice harboring a mutation in the myocardin gene die at embryonic day (E)10.5 of gestation and differentiated vascular SMCs are not observed (14Li S. Wang D.Z. Wang Z. Richardson J.A. Olson E.N. Proc. Natl. Acad. Sci. U. S. A. 2003; 100: 9366-9370Crossref PubMed Scopus (301) Google Scholar). Several important questions related to the role of SRF and myocardin in SMC differentiation remain unanswered. First, is there an alternative factor or pathway that regulates SMC differentiation in the absence of myocardin expression? Of note, in the mouse embryo at E9.5 myocardin is not expressed in the dorsal aorta (at the level of sensitivity afforded by in situ hybridization analyses), but vascular SMCs surrounding the aorta express differentiated markers including SM-α-actin and SM22α (13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar). Second, how do intracellular signals that regulate SMC differentiation, including actin treadmilling and RhoA, modulate activity of SRF and/or myocardin in SMCs? Finally, what role if any do the recently identified myocardin-related transcription factors (MRTFs), MKL1/MAL/BSAC/MRTF-A (henceforth designated MKL1) (19Wang D.Z. Li S. Hockemeyer D. Sutherland L. Wang Z. Schratt G. Richardson J.A. Nordheim A. Olson E.N. Proc. Natl. Acad. Sci. U. S. A. 2002; 99: 14855-14860Crossref PubMed Scopus (405) Google Scholar, 21Ma Z. Morris S.W. Valentine V. Li M. Herbrick J.A. Cui X. Bouman D. Li Y. Mehta P.K. Nizetic D. Kaneko Y. Chan G.C. Chan L.C. Squire J. Scherer S.W. Hitzler J.K. Nat. Genet. 2001; 28: 220-221Crossref PubMed Scopus (246) Google Scholar, 22Mercher T. Coniat M.B. Monni R. Mauchauffe M. Khac F.N. Gressin L. Mugneret F. Leblanc T. Dastugue N. Berger R. Bernard O.A. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 5776-5779Crossref PubMed Scopus (198) Google Scholar, 23Sasazuki T. Sawada T. Sakon S. Kitamura T. Kishi T. Okazaki T. Katano M. Tanaka M. Watanabe M. Yagita H. Okumura K. Nakano H. J. Biol. Chem. 2002; 277: 28853-28860Abstract Full Text Full Text PDF PubMed Scopus (82) Google Scholar) and MRTF-B/MKL2 (19Wang D.Z. Li S. Hockemeyer D. Sutherland L. Wang Z. Schratt G. Richardson J.A. Nordheim A. Olson E.N. Proc. Natl. Acad. Sci. U. S. A. 2002; 99: 14855-14860Crossref PubMed Scopus (405) Google Scholar, 24Selvaraj A. Prywes R. J. Biol. Chem. 2003; 278: 41977-41987Abstract Full Text Full Text PDF PubMed Scopus (114) Google Scholar), play in SMCs and other cell lineages? To better understand the molecular mechanisms that regulate activity of myocardin and SRF in SMCs, we performed a two-hybrid screen in yeast using the conserved N terminus of myocardin as bait. Surprisingly, three clones isolated in this screen encoded MKL1. MKL1 was first identified in the chromosomal translocation t (1Parmacek M.S. Curr. Top. Dev. Biol. 2001; 51: 69-89Crossref PubMed Google Scholar, 22Mercher T. Coniat M.B. Monni R. Mauchauffe M. Khac F.N. Gressin L. Mugneret F. Leblanc T. Dastugue N. Berger R. Bernard O.A. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 5776-5779Crossref PubMed Scopus (198) Google Scholar) (p13;q13) associated with acute megakaryoblastic leukemia in infants and children (21Ma Z. Morris S.W. Valentine V. Li M. Herbrick J.A. Cui X. Bouman D. Li Y. Mehta P.K. Nizetic D. Kaneko Y. Chan G.C. Chan L.C. Squire J. Scherer S.W. Hitzler J.K. Nat. Genet. 2001; 28: 220-221Crossref PubMed Scopus (246) Google Scholar, 22Mercher T. Coniat M.B. Monni R. Mauchauffe M. Khac F.N. Gressin L. Mugneret F. Leblanc T. Dastugue N. Berger R. Bernard O.A. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 5776-5779Crossref PubMed Scopus (198) Google Scholar, 23Sasazuki T. Sawada T. Sakon S. Kitamura T. Kishi T. Okazaki T. Katano M. Tanaka M. Watanabe M. Yagita H. Okumura K. Nakano H. J. Biol. Chem. 2002; 277: 28853-28860Abstract Full Text Full Text PDF PubMed Scopus (82) Google Scholar). Recently, MKL1 has been shown to regulate the transcriptional activity of SRF in fibroblasts, via nuclear translocation in response to serum, RhoA, and actin treadmilling (17Miralles F. Posern G. Zaromytidou A.I. Treisman R. Cell. 2003; 113: 329-342Abstract Full Text Full Text PDF PubMed Scopus (1057) Google Scholar, 25Cen B. Selvaraj A. Burgess R.C. Hitzler J.K. Ma Z. Morris S.W. Prywes R. Mol. Cell. Biol. 2003; 23: 6597-6608Crossref PubMed Scopus (248) Google Scholar). In the studies described in this report, we show that myocardin and MKL1 physically associate via conserved leucine zipper domains. In addition, we demonstrate that MKL1 is expressed in SMCs and examine the signaling pathways regulating nuclear versus cytoplasmic localization of MKL1 in SMCs. Finally, we demonstrated that, like myocardin, MKL1 activates transcriptional regulatory elements controlling SMC-restricted genes and activates endogenous SMC genes in undifferentiated ES cells via SRF-dependent mechanisms. Plasmids—The pcDNA3-MKL1 expression plasmid encoding human MKL1 was generated by subcloning the coding region of the human MKL1 cDNA into pcDNA3 (Invitrogen). The pcDNA3.1-myocardin and pcDNA3-SRF expression plasmids encoding human myocardin and SRF have been described previously (13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar). The pcR3.1-HA-MKL1 and pcDNA3.1-FLAG-MKL1 expression plasmids encoding HA and FLAG epitope-tagged human MKL1, respectively, were generated by subcloning the human MKL1 cDNA into pcR3.1 (Invitrogen) and pcDNA3.1 (Invitrogen), respectively. pcR3.1-myc-myocardin and pcDNA3.1-FLAG-myocardin encoding myc and FLAG epitope-tagged myocardin, respectively, were generated by subcloning the human myocardin cDNA into pcR3.1 (Invitrogen) or pcDNA3.1 (Invitrogen), respectively. The pcDNA-MyoCΔ585 and pcDNA-MyoCΔ381 expression plasmids encoding the N-terminal 585 and 381 amino acids of human myocardin were generated by subcloning each respective myocardin cDNA subfragment into pcDNA3. pcDNA3.1-FLAG-MyoΔLZ expression plasmid is identical to the pcDNA3.1-FLAG myocardin plasmid except the cDNA sequence encoding the myocardin leucine zipper (LZ) domain (amino acids 519–549) was deleted. The p-441.luc reporter plasmid contains the 441-bp mouse SM22α promoter (bp -441 to +41) subcloned into pGL2-Basic (Promega) as described (11Kim S. Ip H.S. Lu M.M. Clendenin C. Parmacek M.S. Mol. Cell. Biol. 1997; : 2266-2278Crossref PubMed Scopus (190) Google Scholar). The p-441μCArG. luc reporter is identical to the -441.luc reporter plasmid except that the underlined nucleotides in SME-1 (5′-TCCTGCCCATAGATCTTTTTTCC-3′) and SME-4 (5′-GCTCCAACTTGGTGTCTTTCCCCGGATATGGAGCCT-3′) were mutated to abolish SRF binding (11Kim S. Ip H.S. Lu M.M. Clendenin C. Parmacek M.S. Mol. Cell. Biol. 1997; : 2266-2278Crossref PubMed Scopus (190) Google Scholar). pSME4-luc contains four copies of the mouse SM22α SME-4 CArG box-containing nuclear protein binding site (bp -173 to -136) subcloned immediately 5′ of the minimal promoter in pLuc-MCS (Stratagene) (26Strobeck M. Kim S. Zhang J.C. Clendenin C. Du K.L. Parmacek M.S. J. Biol. Chem. 2001; 276: 16418-16424Abstract Full Text Full Text PDF PubMed Scopus (38) Google Scholar). The pPI-Act.luc and pPI-MHC.luc luciferase reporter plasmids containing the SM-α-actin and SM-MHC promoter and intragenic enhancers, respectively, subcloned into pGL3-Basic, were the generous gift of Dr. Gary Owens (University of Virginia) and have been described previously (13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar). pSRE-luc contains four copies of the c-fos SRE subcloned immediately 5′ of the minimal promoter pLuc-MCS. pCRE-luc contains four copies of a consensus cyclic AMP response element (CRE) (27Lee K.A. Masson N. Biochim. Biophys. Acta. 1993; 1174: 221-233Crossref PubMed Scopus (189) Google Scholar), subcloned 5′ of the minimal promoter in pLuc-MCS. pGBKT7-MyoCΔ585 encodes the N-terminal 585 amino acid residues of myocardin, subcloned in-frame to the GAL4 binding domain of pGBKT7 (Clontech). The pRK5-myc-RhoA N19 expression plasmid encodes a dominant-negative RhoA mutant and was a generous gift from Professor Alan Hall (University College London). Yeast Two-hybrid Screen—pGBKT7-MyoCΔ585 was transformed into yeast strain AH109 to generate the bait strain. >5 × 109 of the bait strain cells were mated with >1.6 × 108 colony forming units of the pre-transformed Human Heart MATCHMAKER library (Clontech) and plated onto SD-Ade/-His/-Leu/-Trp/+X-α-Gal agar plates. Positive clones were identified by growth on SD-Ade/-His/-Leu/-Trp/+X-α-Gal agar plates. 5.1 × 105 transformants were screened based on colony size and intensity of X-Gal staining, and plasmid was recovered from the colonies and sequenced as recommended by the manufacturer (Clontech). Three clones corresponding to the full-length open reading frame of MKL1 were isolated and characterized further. Co-immunoprecipitation Assays—In vitro transcription reactions and translations were performed using pcDNA-MyoCΔ585, pcDNA-MyoCΔ381, pcDNA3.1-MyoΔLZ, and pcDNA3.1-FLAG-MKL1, respectively, as templates and the TnT Quick Coupled Transcription/Translation System (Promega). Myocardin and each deletion mutant were labeled with biotinylated-lysyl-tRNA (Transcend tRNA, Promega). Unlabeled FLAG-MKL1, biotinylated myocardin, and 40 μl of EZview Red anti-FLAG M2 affinity gel (Sigma) were incubated at 4 °C for 12–16 h. Protein bound to the anti-FLAG M2 antibody was isolated and the immunoprecipitated protein was electrophoretically separated by SDS-PAGE, transferred to polyvinylidene difluoride membrane, and visualized with streptavidin-horseradish peroxidase and a chemiluminescent substrate (Promega). Northern Blot Analyses—Northern blot analyses were performed using the Human 12-lane MTN, Human Muscle MTN, and Human Cardiovascular MTN blots prepared by Clontech as described previously (28Parmacek M.S. Leiden J.M. J. Biol. Chem. 1989; 264: 13217-13225Abstract Full Text PDF PubMed Google Scholar). In addition, Northern blot analyses were performed on membranes containing 10 μg/lane of total RNA harvested from wild-type ES cells, NIH3T3 cells, C3H10T1/2 cells, C2C12 myoblasts, A10 SMCs, PAC1 SMCs, and primary rat aortic SMCs, respectively, as described previously (28Parmacek M.S. Leiden J.M. J. Biol. Chem. 1989; 264: 13217-13225Abstract Full Text PDF PubMed Google Scholar). The 32P-labeled human and mouse MKL1 cDNA probes were generated by random primed labeling as described previously (28Parmacek M.S. Leiden J.M. J. Biol. Chem. 1989; 264: 13217-13225Abstract Full Text PDF PubMed Google Scholar). Cell Culture, Transient Co-transfections, and Luciferase Assays— COS-7 cells were grown as described previously (13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar). Transient co-transfection of 1 × 105 COS-7 cells were performed with FuGENE 6 (Roche Diagnostics) and 200 ng to 1 μg of the indicated luciferase reporter plasmid, 100–200 ng of the indicated expression plasmid, and 10 ng of the phRL-TK (-Int) reference plasmid (Promega). Luciferase activity was measured and normalized for transfection efficiency using the Dual Luciferase Assay system (Promega). Data are reported as mean normalized relative light units ± S.E. SRF-/- ES cells (13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar) were grown and transfected with LipofectAMINE 2000 (Invitrogen) as described previously (13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar). NIH3T3 cells, A7r5 SMCs, C3H10T1/2 cells, A10 SMCs, and PAC1 SMCs ATCC were grown as described previously (29Solway J. Seltzer J. Samaha F.F. Kim S. Alger L.E. Niu Q. Morrisey E.E. Ip H.S. Parmacek M.S. J. Biol. Chem. 1995; 270: 13460-13469Abstract Full Text Full Text PDF PubMed Scopus (227) Google Scholar). Transfection of ES Cells and Real Time Reverse Transcriptase (RT)-PCR—1 × 106 SRF-/- ES cells (13Du K.L. Ip H.S. Li J. Chen M. Dandre F. Yu W. Lu M.M. Owens G.K. Parmacek M.S. Mol. Cell. Biol. 2003; 23: 2425-2437Crossref PubMed Scopus (312) Google Scholar) were co-transfected with LipofectAMINE 2000 (Invitrogen) and either 4 μg of pcDNA3-myocardin plus 10 ng of pcDNA3-SRF, 4 μg of pcDNA3-MKL1 plus 10 ng of pcDNA3-SRF, or 4 μg of pcDNA3 plus 10 ng of pcDNA3-SRF. 24 h post-transfection, RNA was harvested from the cells for real time RT-PCR with TRIzol (Invitrogen). PCR were performed with 5 μl of reverse transcribed cDNA reaction mixture, 400 nm of specific forward and reverse primers and 1× SYBR Green PCR Master Mix (Applied Biosystems). The following primer pairs were utilized: (i) SM-α-actin, 5′-GAGAAGCCCAGCCAGTCG-3′ and 5′-CTCTTGCTCTGGGCTTCA-3′; (ii) myocardin, 5′-CTGTGTGGAGTCCTCAGGTCAAACC-3′ and 5′-GATGTGCTGCGGGCTCTTCAG-3′; (iii) calponin-h1, 5′-CCCAGAAATACGACCATCAGCG-3′ and 5′-CACCCCCTCAATCCACTCTCTCAG-3′; (iv) SM22α, 5′-AGCCAGTGAAGGTGCCTGAGAAC-3′ and 5′-TGCCCAAAGCCATTAGAGTCCTC-3′; (v) SM-MHC, 5′-TGGACACCATGTCAGGGAAA-3′ and 5′-ATGGACACAAGTGCTAAGCAGTCT-3′; (vi) glyceraldehyde-3-phosphate dehydrogenase, 5′-GTGGCAAAGTGGAGATTGTTGCC-3′ and 5′-GATGATGACCCGTTTGGCTCC-3′. Quantification of the reaction product was performed using the MJ Research DNA Engine Opticon 2 real time detection system. PCR cycle conditions were 95 °C for 10 min, followed by 40 cycles of denaturation at 95 °C for 15 s and annealing and extension at 60 °C for 1 min. All RT and PCR were performed in triplicate with and without RT as controls. Cycle threshold (C(t)) values were converted to relative gene expression levels using the 2-ΔΔC(t) method (30Livak K.J. Schmittgen T.D. Methods (Orlando). 2001; 25: 402-408Crossref Scopus (124899) Google Scholar). Chromatin Immunoprecipitation (ChIP) Assays—ChIP assays were performed using the protocol described by Owens and colleagues (31Manabe I. Owens G.K. Circ. Res. 2001; 88: 1127-1134Crossref PubMed Scopus (143) Google Scholar). SRF-/- ES cells were transiently transfected with pcDNA3.1, pcDNA3.1-FLAG-Myo, pcDNA3.1-FLAG-MyoΔLZ, pcDNA3.1-FLAG-Myo plus pcDNA3-SRF, or pcDNA3.1-FLAG-MyoΔLZ plus pcDNA3-SRF, respectively. 24 h post-transfection, cells were fixed in 1% formaldehyde, lysed in 1% SDS, 10 mm EDTA, and 50 mm Tris-HCl, pH 8.1, with mammalian proteinase inhibitors (Sigma) and sonicated. Each cell lysate was diluted 10-fold, incubated overnight with anti-FLAG M2 antibody (Sigma), and 90 μg of Protein A-agarose was added to each sample. Immunoprecipitated, reverse cross-linked DNA was recovered by phenol/chloroform extraction and in turn was subject to PCR analysis with primers flanking the SME-4 CArG box containing the nuclear protein binding site in the mouse SM22α promoter (5′-GGTCCTGCCCATAAAAGGTTT-3′ and 5′-TGCCCATGGAAGTCTGCTTGG-3′) designed to generate a 200-bp amplified DNA fragment. Immunohistochemistry—The intracellular location of myocardin and MKL1 was determined by transiently co-transfecting primary rat aortic SMCs, A7r5 SMCs, and NIH3T3 cells with 1 μg of pcR3.1-HA-MKL, pcR3.1-myc-myocardin, or the control pcDNA3 expression vector with FuGENE 6 (Roche). 24 h post-transfection cells were serum-starved for 48 h in Dulbecco's modified Eagle's medium supplemented with 0.1% fetal bovine serum. Subsequently, the cells were either maintained in 0.1% fetal bovine serum or serum-stimulated with Dulbecco's modified Eagle's medium supplemented with 20% fetal bovine serum for 1 h. To determine the effect of actin polymerization on the intracellular localization of MKL1, cells were treated for 1 h with 0.5 × 10-6m latrunculin B (Alexis), which inhibits polymerization of actin filaments (32Zhang J.C. Helmke B.P. Shum A. Du K. Yu W.W. Lu M.M. Davies P.F. Parmacek M.S. Mech. Dev. 2002; 115: 161-166Crossref PubMed Scopus (26) Google Scholar). To promote actin polymerization, cells were treated for 1 h with 0.5 × 10-6m jasplakinolide (Molecular Probes) (32Zhang J.C. Helmke B.P. Shum A. Du K. Yu W.W. Lu M.M. Davies P.F. Parmacek M.S. Mech. Dev. 2002; 115: 161-166Crossref PubMed Scopus (26) Google Scholar). To determine the effect of RhoA signaling on the intracellular localization of MKL1, cells were transfected with the pRK5-myc-RhoA N19 expression plasmid encoding a dominant-negative RhoA mutant protein. The cells were then fixed, permeabilized, incubated with primary and secondary antibodies, and visualized on a Zeiss Axiophot microscope as described previously (32Zhang J.C. Helmke B.P. Shum A. Du K. Yu W.W. Lu M.M. Davies P.F. Parmacek M.S. Mech. Dev. 2002; 115: 161-166Crossref PubMed Scopus (26) Google Scholar). MKL1 Physically Associates with Myocardin—To examine the molecular mechanisms regulating myocardin activity, a yeast two-hybrid screen of a human heart cDNA yeast expression library was performed using a yeast indicator strain expressing the N terminus (amino acids 1–585) of the human myocardin protein as bait. The N terminus of myocardin was utilized because this region of the protein includes the evolutionarily conserved and functionally important RPEL, glutamine-rich, basic and SAP domains that have been implicated in regulating activity of myocardin (Fig. 1A and Ref. 18Wang D. Chang P.S. Wang Z. Sutherland L. Richardson J.A. Small E. Krieg P.A. Olson E.N. Cell. 2001; 105: 851-862Abstract Full Text Full Text PDF PubMed Scopus (752) Google Scholar). Positive clones were identified by growth on SD-Ade/-His/-Leu/-Trp/+X-α-Gal agar plates. Three of the 58 isolates contained an ∼4-kb cDNA insert that corresponded to the full-length coding region (open reading frame) of human MKL1/MAL/MRTF-A protein (19Wang D.Z. Li S. Hockemeyer D. Sutherland L. Wang Z. Schratt G. Richardson J.A. Nordheim A. Olson E.N. Proc. Natl. Acad. Sci. U. S. A. 2002; 99: 14855-14860Crossref PubMed Scopus (405) Google Scholar, 21Ma Z. Morris S.W. Valentine V. Li M. Herbrick J.A. Cui X. Bouman D. Li Y. Mehta P.K. Nizetic D. Kaneko Y. Chan G.C. Chan L.C. Squire J. Scherer S.W. Hitzler J.K. Nat. Genet. 2001; 28: 220-221Crossref PubMed Scopus (246) Google Scholar, 22Mercher T. Coniat M.B. Monni R. Mauchauffe M. Khac F.N. Gressin L. Mugneret F. Leblanc T. Dastugue N. Berger R. Bernard O.A. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 5776-5779Crossref PubMed Scopus (198) Google Scholar). The coding regions of these three clones were identical with differences observed only in the extent of the 5′- and 3′-untranslated regions. A BLAST search of GenBank™ revealed that the human MKL1 gene contains 15 exons spanning 226 kb of sequence on chromosome 1. The 3,907-bp human MKL1 cDNA encodes a 98.9-kDa protein. Sequence analysis revealed that the deduced human MKL1 protein shares 33% homology with myocardin over the entire coding sequence and 70% homology with myocardin across its N-terminal region containing the RPEL, basic, glutamine-rich, SAP, and leucine zipper domains (Fig. 1A). Of note, MKL1
What problem does this paper attempt to address?
-
The Long Noncoding RNA Cardiac Mesoderm Enhancer- Associated Noncoding RNA (carmn) is a Critical Regulator of Gastrointestinal Smooth Muscle Contractile Function and Motility
Xiangqin He,Kunzhe Dong,Jian Shen,Guoqing Hu,James D. Mintz,Reem T. Atawia,Juanjuan Zhao,Xiuxu Chen,Robert W. Caldwell,Meixiang Xiang,David W. Stepp,David J. Fulton,Jiliang Zhou
DOI: https://doi.org/10.1053/j.gastro.2023.03.229
IF: 29.4
2023-01-01
Gastroenterology
Abstract:BACKGROUND & AIMS: Visceral smooth muscle cells (SMCs) are an integral component of the gastrointestinal (GI) tract that regulate GI motility. SMC contraction is regulated by post-translational signaling and the state of differentiation. Impaired SMC contraction is associated with significant morbidity and mortality, but the mechanisms regulating SMC-specific contractile gene expression, including the role of long noncoding RNAs (lncRNAs), remain largely unexplored. Herein, we reveal a critical role of Carmn (cardiac mesoderm enhancer-associated noncoding RNA), an SMC-specific lncRNA, in regulating visceral SMC phenotype and contractility of the GI tract. METHODS: Genotype-Tissue Expression and publicly available single-cell RNA sequencing (scRNA-seq) data sets from embryonic, adult human, and mouse GI tissues were interrogated to identify SMC-specific lncRNAs. The functional role of Carmn was investigated using novel green fluorescent protein (GFP) knock-in (KI) reporter/knock-out (KO) mice. Bulk RNA-seq and single nucleus RNA sequencing (snRNA-seq) of colonic muscularis were used to investigate underlying mechanisms. RESULTS: Unbiased in silico analyses and GFP expression patterns in Carmn GFP KI mice revealed that Carmn is highly expressed in GI SMCs in humans and mice. Premature lethality was observed in global Carmn KO and inducible SMC-specific KO mice due to GI pseudo-obstruction and severe distension of the GI tract, with dysmotility in cecum and colon segments. Histology, GI transit, and muscle myography analysis revealed severe dilation, significantly delayed GI transit, and impaired GI contractility in Carmn KO vs control mice. Bulk RNA-seq of GI muscularis revealed that loss of Carmn promotes SMC pheno-typic switching, as evidenced by up-regulation of extracellular matrix genes and down-regulation of SMC contractile genes, including Mylk, a key regulator of SMC contraction. snRNA-seq further revealed SMC Carmn KO not only compromised myogenic motility by reducing contractile gene expression but also impaired neurogenic motility by disrupting cell-cell con-nectivity in the colonic muscularis. These findings may have translational significance, because silencing CARMN in human colonic SMCs significantly attenuated contractile gene expres-sion, including MYLK, and decreased SMC contractility. Lucif-erase reporter assays showed that CARMN enhances the transactivation activity of the master regulator of SMC contractile phenotype, myocardin, thereby maintaining the GI SMC myogenic program. CONCLUSIONS: Our data suggest that Carmn is indispensable for maintaining GI SMC contractile function in mice and that loss of function of CARMN may contribute to human visceral myopathy. To our knowledge this is the first study showing an essential role of lncRNA in the regulation of visceral SMC phenotype.
-
CARMN is an Evolutionarily Conserved Smooth Muscle Cell-Specific LncRNA That Maintains Contractile Phenotype by Binding Myocardin
Kunzhe Dong,Jian Shen,Xiangqin He,Guoqing Hu,Liang Wang,Islam Osman,Kristopher M. Bunting,Rachael Dixon-Melvin,Zeqi Zheng,Hongbo Xin,Meixiang Xiang,Almira Vazdarjanova,David J. R. Fulton,Jiliang Zhou
DOI: https://doi.org/10.1161/circulationaha.121.055949
IF: 37.8
2021-01-01
Circulation
Abstract:BACKGROUND:Vascular homeostasis is maintained by the differentiated phenotype of vascular smooth muscle cells (VSMCs). The landscape of protein coding genes comprising the transcriptome of differentiated VSMCs has been intensively investigated but many gaps remain including the emerging roles of noncoding genes.METHODS:We reanalyzed large-scale, publicly available bulk and single-cell RNA sequencing datasets from multiple tissues and cell types to identify VSMC-enriched long noncoding RNAs. The in vivo expression pattern of a novel smooth muscle cell (SMC)-expressed long noncoding RNA, Carmn (cardiac mesoderm enhancer-associated noncoding RNA), was investigated using a novel Carmn green fluorescent protein knock-in reporter mouse model. Bioinformatics and quantitative real-time polymerase chain reaction analysis were used to assess CARMN expression changes during VSMC phenotypic modulation in human and murine vascular disease models. In vitro, functional assays were performed by knocking down CARMN with antisense oligonucleotides and overexpressing Carmn by adenovirus in human coronary artery SMCs. Carotid artery injury was performed in SMC-specific Carmn knockout mice to assess neointima formation and the therapeutic potential of reversing CARMN loss was tested in a rat carotid artery balloon injury model. The molecular mechanisms underlying CARMN function were investigated using RNA pull-down, RNA immunoprecipitation, and luciferase reporter assays.RESULTS:We identified CARMN, which was initially annotated as the host gene of the MIR143/145 cluster and recently reported to play a role in cardiac differentiation, as a highly abundant and conserved, SMC-specific long noncoding RNA. Analysis of the Carmn GFP knock-in mouse model confirmed that Carmn is transiently expressed in embryonic cardiomyocytes and thereafter becomes restricted to SMCs. We also found that Carmn is transcribed independently of Mir143/145. CARMN expression is dramatically decreased by vascular disease in humans and murine models and regulates the contractile phenotype of VSMCs in vitro. In vivo, SMC-specific deletion of Carmn significantly exacerbated, whereas overexpression of Carmn markedly attenuated, injury-induced neointima formation in mouse and rat, respectively. Mechanistically, we found that Carmn physically binds to the key transcriptional cofactor myocardin, facilitating its activity and thereby maintaining the contractile phenotype of VSMCs.CONCLUSIONS:CARMN is an evolutionarily conserved SMC-specific long noncoding RNA with a previously unappreciated role in maintaining the contractile phenotype of VSMCs and is the first noncoding RNA discovered to interact with myocardin.
-
NG2/CSPG4, CD146/MCAM and VAP1/AOC3 Are Regulated by Myocardin-Related Transcription Factors in Smooth Muscle Cells
Catarina Rippe,Björn Morén,Li Liu,Karin G. Stenkula,Johan Mustaniemi,Malin Wennström,Karl Swärd
DOI: https://doi.org/10.1038/s41598-021-85335-x
IF: 4.6
2021-01-01
Scientific Reports
Abstract:The present work addressed the hypothesis that NG2/ CSPG4 , CD146/ MCAM , and VAP1/ AOC3 are target genes of myocardin-related transcription factors (MRTFs: myocardin/ MYOCD , MRTF-A/ MKL1 , MRTF-B/ MKL2 ) and serum response factor ( SRF ). Using a bioinformatics approach, we found that CSPG4 , MCAM , and AOC3 correlate with MYOCD , MRTF-A/ MKL1 , and SRF across human tissues. No other transcription factor correlated as strongly with these transcripts as SRF . Overexpression of MRTFs increased both mRNA and protein levels of CSPG4 , MCAM , and AOC3 in cultured human smooth muscle cells (SMCs). Imaging confirmed increased staining for CSPG4, MCAM, and AOC3 in MRTF-A/ MKL1 -transduced cells. MRTFs exert their effects through SRF, and the MCAM and AOC3 gene loci contained binding sites for SRF. SRF silencing reduced the transcript levels of these genes, and time-courses of induction paralleled the direct target ACTA2 . MRTF-A/ MKL1 increased the activity of promoter reporters for MCAM and AOC3 , and transcriptional activation further depended on the chromatin remodeling enzyme KDM3A. CSPG4 , MCAM , and AOC3 responded to the MRTF-SRF inhibitor CCG-1423, to actin dynamics, and to ternary complex factors. Coincidental detection of these proteins should reflect MRTF-SRF activity, and beyond SMCs, we observed co-expression of CD146/ MCAM , NG2/ CSPG4 , and VAP1/ AOC3 in pericytes and endothelial cells in the human brain. This work identifies highly responsive vascular target genes of MRTF-SRF signaling that are regulated via a mechanism involving KDM3A.
-
Regulation of the Muscarinic M3 Receptor by Myocardin-Related Transcription Factors
Li Liu,Catarina Rippe,Ola Hansson,Dmytro Kryvokhyzha,Steven Fisher,Mari Ekman,Karl Swärd
DOI: https://doi.org/10.3389/fphys.2021.710968
IF: 4
2021-09-03
Frontiers in Physiology
Abstract:Myocardin-related transcription factors (MRTFs: myocardin/ MYOCD , MRTF-A/ MRTFA , and MRTF-B/ MRTFB ) are co-factors of serum response factor (SRF) that activate the smooth muscle cell (SMC) gene program and that play roles in cardiovascular development and mechanobiology. Gain and loss of function experiments have defined the SMC gene program under control of MRTFs, yet full understanding of their impact is lacking. In the present study, we tested the hypothesis that the muscarinic M 3 receptor ( CHRM3 ) is regulated by MRTFs together with SRF. Forced expression of MYOCD (8d) in human coronary artery (SMC) followed by RNA-sequencing showed increased levels of M 2 , M 3 , and M 5 receptors ( CHRM2 : 2-fold, CHRM3 : 16-fold, and CHRM5 : 2-fold). The effect of MYOCD on M 3 was confirmed by RT-qPCR using both coronary artery and urinary bladder SMCs, and correlation analyses using human transcriptomic datasets suggested that M 3 may also be regulated by MRTF-B. Head-to-head comparisons of MYOCD, MRTF-A and MRTF-B, argued that while all MRTFs are effective, MRTF-B is the most powerful transactivator of CHRM3 , causing a 600-fold increase at 120h. Accordingly, MRTF-B conferred responsiveness to the muscarinic agonist carbachol in Ca 2+ imaging experiments. M 3 was suppressed on treatment with the MRTF-SRF inhibitor CCG-1423 using SMCs transduced with either MRTF-A or MRTF-B and using intact mouse esophagus in culture (by 92±2%). Moreover, silencing of SRF with a short hairpin reduced CHRM3 (by >60%) in parallel with α-actin ( ACTA2 ). Tamoxifen inducible knockout of Srf in smooth muscle reduced Srf (by 54±4%) and Chrm3 (by 41±6%) in the urinary bladder at 10days, but Srf was much less reduced or unchanged in aorta, ileum, colon, trachea, and esophagus. Longer induction (21d) further accentuated the reduction of Chrm3 in the bladder and ileum, but no change was seen in the aorta. Single cell RNA-sequencing revealed that Mrtfb dominates in ECs, while Myocd dominates in SMCs, raising the possibility that Chrm3 may be driven by Mrtfb-Srf in the endothelium and by Myocd-Srf in SMCs. These findings define a novel transcriptional control mechanism for muscarinic M 3 receptors in human cells, and in mice, that could be targeted for therapy.
physiology
-
Myocardin regulates exon usage in smooth muscle cells through induction of splicing regulatory factors
Li Liu,Dmytro Kryvokhyzha,Catarina Rippe,Aishwarya Jacob,Andrea Borreguero-Muñoz,Karin G. Stenkula,Ola Hansson,Christopher W. J. Smith,Steven A. Fisher,Karl Swärd
DOI: https://doi.org/10.1007/s00018-022-04497-7
IF: 8
2022-08-02
Cellular and Molecular Life Sciences
Abstract:Differentiation of smooth muscle cells (SMCs) depends on serum response factor (SRF) and its co-activator myocardin ( MYOCD ). The role of MYOCD for the SMC program of gene transcription is well established. In contrast, the role of MYOCD in control of SMC-specific alternative exon usage, including exon splicing, has not been explored. In the current work we identified four splicing factors ( MBNL1 , RBPMS , RBPMS2 , and RBFOX2 ) that correlate with MYOCD across human SMC tissues. Forced expression of MYOCD family members in human coronary artery SMCs in vitro upregulated expression of these splicing factors. For global profiling of transcript diversity, we performed RNA-sequencing after MYOCD transduction. We analyzed alternative transcripts with three different methods. Exon-based analysis identified 1637 features with differential exon usage. For example, usage of 3 ́ exons in MYLK that encode telokin increased relative to 5 ́ exons, as did the 17 kDa telokin to 130 kDa MYLK protein ratio. Dedicated event-based analysis identified 239 MYOCD-driven splicing events. Events involving MBNL1 , MCAM , and ACTN1 were among the most prominent, and this was confirmed using variant-specific PCR analyses. In support of a role for RBPMS and RBFOX2 in MYOCD-driven splicing we found enrichment of their binding motifs around differentially spliced exons. Moreover, knockdown of either RBPMS or RBFOX2 antagonized splicing events stimulated by MYOCD, including those involving ACTN1 , VCL , and MBNL1 . Supporting an in vivo role of MYOCD-SRF-driven splicing, we demonstrate altered Rbpms expression and splicing in inducible and SMC-specific Srf knockout mice. We conclude that MYOCD-SRF, in part via RBPMS and RBFOX2, induce a program of differential exon usage and alternative splicing as part of the broader program of SMC differentiation.
cell biology,biochemistry & molecular biology
-
Rho-actin signaling to the MRTF coactivators dominates the immediate transcriptional response to serum in fibroblasts
Cyril Esnault,Aengus Stewart,Francesco Gualdrini,Phil East,Stuart Horswell,Nik Matthews,Richard Treisman
DOI: https://doi.org/10.1101/gad.239327.114
2014-05-01
Abstract:The transcription factor SRF (serum response factor) recruits two families of coactivators, the MRTFs (myocardin-related transcription factors) and the TCFs (ternary complex factors), to couple gene transcription to growth factor signaling. Here we investigated the role of the SRF network in the immediate transcriptional response of fibroblasts to serum stimulation. SRF recruited its cofactors in a gene-specific manner, and virtually all MRTF binding was directed by SRF. Much of SRF DNA binding was serum-inducible, reflecting a requirement for MRTF-SRF complex formation in nucleosome displacement. We identified 960 serum-responsive SRF target genes, which were mostly MRTF-controlled, as assessed by MRTF chromatin immunoprecipitation (ChIP) combined with deep sequencing (ChIP-seq) and/or sensitivity to MRTF-linked signals. MRTF activation facilitates RNA polymerase II (Pol II) recruitment or promoter escape according to gene context. MRTF targets encode regulators of the cytoskeleton, transcription, and cell growth, underpinning the role of SRF in cytoskeletal dynamics and mechanosensing. Finally, we show that specific activation of either MRTFs or TCFs can reset the circadian clock.
-
Microrna-34a Regulated Sirtuin 1 Is Required For Smooth Muscle Cell Differentiation From Murine And Human Embryonic Stem Cells
Xiaotian Yu,Li Zhang,Guanmei Wen,Hanqing Zhao,Yuan Huang,Jianhua Zhu,Qingbo Xu,Shu Ye,Wen Wang,Qingzhong Xiao
IF: 37.8
2013-01-01
Circulation
Abstract:Background: microRNA-34a (miR-34a) has been recently reported to regulate tumor cell cycle progression and apoptosis. However, very little is known about the functional role of miR-34a in smooth muscle cell (SMC) differentiation from stem cells. In the present study, we assessed the hypothesis that miR34a and its target genes play an important role in SMC differentiation. Methods and results: Mouse embryonic stem (ES) cells were seeded on collagen IV-coated flasks and cultured in differentiation medium for 4 to 8 days to allow for SMC differentiation. miR-34a was significantly upregulated during SMC differentiation. Enforced expression and knockdown of miR34a in differentiating ES cells significantly promoted and inhibited SMC-specific gene expressions, respectively. Furthermore, overexpression and knockdown of miR-34a upregulated and downregulated several SMC transcription factors, including SRF, myocardin and MEF2c in a similar manner, and miR-34a overexpression in stem cells promoted SMC differentiatio...
-
Cell Type Dependent Suppression of Inflammatory Mediators by Myocardin Related Transcription Factors
Li Liu,Elisabeth Bankell,Catarina Rippe,Bjorn Moren,Karin G. Stenkula,Bengt-Olof Nilsson,Karl Sward
DOI: https://doi.org/10.3389/fphys.2021.732564
IF: 4
2021-01-01
Frontiers in Physiology
Abstract:Myocardin related transcription factors (MRTFs: MYOCD/myocardin, MRTF-A, and MRTF-B) play a key role in smooth muscle cell differentiation by activating contractile genes. In atherosclerosis, MRTF levels change, and most notable is a fall of MYOCD. Previous work described anti-inflammatory properties of MRTF-A and MYOCD, occurring through RelA binding, suggesting that MYOCD reduction could contribute to vascular inflammation. Recent studies have muddled this picture showing that MRTFs may show both anti- and pro-inflammatory properties, but the basis of these discrepancies remain unclear. Moreover, the impact of MRTFs on inflammatory signaling pathways in tissues relevant to human arterial disease is uncertain. The current work aimed to address these issues. RNA-sequencing after forced expression of myocardin in human coronary artery smooth muscle cells (hCASMCs) showed reduction of pro-inflammatory transcripts, including CCL2, CXCL8, IL6, and IL1B. Side-by-side comparison of MYOCD, MRTF-A, and MRTF-B in hCASMCs, showed that the anti-inflammatory impact was shared among MRTFs. Correlation analyses using human arterial transcriptomic datasets revealed negative correlations between MYOCD, MRTFA, and SRF, on the one hand, and the inflammatory transcripts, on the other. A pro-inflammatory drive from lipopolysaccharide, did not change the size of the suppressive effect of MRTF-A in hCASMCs on either mRNA or protein levels. To examine cell type-dependence, we compared the anti-inflammatory impact in hCASMCs, with that in human bladder SMCs, in endothelial cells, and in monocytes (THP-1 cells). Surprisingly, little anti-inflammatory activity was seen in endothelial cells and monocytes, and in bladder SMCs, MRTF-A was pro-inflammatory. CXCL8, IL6, and IL1B were increased by the MRTF-SRF inhibitor CCG-1423 and by MRTF-A silencing in hCASMCs, but depolymerization of actin, known to inhibit MRTF activity, had no stimulatory effect, an exception being IL1B. Co-immunoprecipitation supported binding of MRTF-A to RelA, supporting sequestration of this important pro-inflammatory mediator as a mechanism. Dexamethasone treatment and silencing of RelA (by 76 +/- 1%) however only eliminated a fraction of the MRTF-A effect (approximate to 25%), suggesting mechanisms beyond RelA binding. Indeed, SRF silencing suggested that MRTF-A suppression of IL1B and CXCL8 depends on SRF. This work thus supports an anti-inflammatory impact of MRTF-SRF signaling in hCASMCs and in intact human arteries, but not in several other cell types.
-
Abstract 11470: Functional Validation of the Novel Role of SMARCB1 in Myocardial Fibrosis
Min WangMahsima ShabaniDuan LiuJyotan SahniHazley J PyfferoenJoao A Ac LimaNaveen L PereiraMayo Clicnic,Rochester,MNBaltimore,MDDept of Molecular Pharmacology and Experimental Therapeutics,Mayo Clicnic,Rochester,MNJOHNS HOPKINS UNIVERSITY,Baltimore,MDMayo Clinic,Rochester,MN
DOI: https://doi.org/10.1161/circ.146.suppl_1.11470
IF: 37.8
2022-11-01
Circulation
Abstract:Circulation, Volume 146, Issue Suppl_1, Page A11470-A11470, November 8, 2022. Introduction:It has been previously demonstrated that cardiomyopathy predisposing common genetic variation inSMARCB1is associated with interstitial myocardial fibrosis (MF) as determined by cardiac MRI (CMR) linking the development of cardiomyopathy with MF.SMARCB1is known to be a tumor suppressor, but it's role in the pathophysiology of MF and fibroblast physiology has not been functionally validated.Hypothesis:SMARCB1plays an important role in regulating human cardiac fibroblast (HCF) differentiation/activation and collagen deposition, which are the critical steps in the development of MF.Methods:Cultured HCFs were treated with TGF-β1 (10ng/ml) for 48hrs to transdifferentiate into myofibroblasts, which are characterized by increased expression of α-smooth muscle actin (α-SMA, ACTA2) and collagen I (COL1A1). HCFs were transfected by siRNA using LipofectamineTM RNAiMAX Transfection Reagent to knock-down (KD) the expression ofSMARCB1. Total RNA and protein were extracted from cell lysate followed by qPCR and Western blot (WB) to quantify the expression level of SMARCB1, ACTA2 and COL1A1. Triplicate experiments were performed using biologically independent samples. For statistical analysis, a paired two-tail t-test was used.Results:HCFs treatment with TGF-β1, resulted in a significant decrease in expression ofSMARCB1(p=0.031) with accompanying significant increase in expression of ACTA2 (p=0.025) and COL1A1 (p=0.0015) as compared to control by qPCR and WB.SMARCB1KD resulted in a further significant increase in expression of ACTA2 (p=0.018) and COL1A1 (p=0.028) as compared to non-target siRNA control group at both the transcript and protein level.Conclusion:We demonstrate for the first time that a gene associated with the development of cardiomyopathySMARCB1mediates TGF-β1 induced cardiac fibrosis. These findings suggestSMARCB1as a potential therapeutic target to attenuate MF in cardiomyopathy.
cardiac & cardiovascular systems,peripheral vascular disease
-
p90RSK2, a new MLCK mediates contractility in myosin light chain kinase null smooth muscle
Jaspreet Kalra,Mykhaylo Artamonov,Hua Wang,Aaron Franke,Zaneta Markowska,Li Jin,Zygmunt S Derewenda,Ramon J Ayon,Avril Somlyo
DOI: https://doi.org/10.3389/fphys.2023.1228488
2023-09-13
Abstract:Introduction: Phosphorylation of smooth muscle (SM) myosin regulatory light chain (RLC20) is a critical switch leading to SM contraction. The canonical view held that only the short isoform of myosin light chain kinase (MLCK1) catalyzed this reaction. It is now accepted that auxiliary kinases may contribute to vascular SM tone and contractility. We have previously reported that p90 ribosomal S6 kinase (RSK2) functions as such a kinase, in parallel with MLCK1, contributing ∼25% of the maximal myogenic force in resistance arteries. Thus, RSK2 may be instrumental in the regulation of basal vascular tone and blood pressure. Here, we take advantage of a MLCK1 null mouse (mylk1 -/-) to further test our hypothesis that RSK2 can function as an MLCK, playing a significant physiological role in SM contractility. Methods: Using fetal (E14.5-18.5) SM tissues, as embryos die at birth, we investigated the necessity of MLCK for contractility and fetal development and determined the ability of RSK2 kinase to compensate for the lack of MLCK and characterized its signaling pathway in SM. Results and Discussion: Agonists induced contraction and RLC20 phosphorylation in mylk1 -/- SM was attenuated by RSK2 inhibition. The pCa-tension relationships in permeabilized strips of bladder showed no difference in Ca2+ sensitivity in WT vs mylk1 -/- muscles, although the magnitude of force responses was considerably smaller in the absence of MLCK. The magnitude of contractile responses was similar upon addition of GTPγS to activate the RhoA/ROCK pathway or calyculinA to inhibit the myosin phosphatase. The Ca2+-dependent tyrosine kinase, Pyk2, contributed to RSK2-mediated contractility and RLC20 phosphorylation. Proximity-ligation and immunoprecipitation assays demonstrated an association of RSK2, PDK1 and ERK1/2 with MLCK and actin. RSK2, PDK1, ERK1/2 and MLCK formed a signaling complex on the actin filament, positioning them for interaction with adjacent myosin heads. The Ca2+-dependent component reflected the agonist mediated increases in Ca2+, which activated the Pyk2/PDK1/RSK2 signaling cascade. The Ca2+-independent component was through activation of Erk1/2/PDK1/RSK2 leading to direct phosphorylation of RLC20, to increase contraction. Overall, RSK2 signaling constitutes a new third signaling pathway, in addition to the established Ca2+/CaM/MLCK and RhoA/ROCK pathways to regulate SM contractility.
-
Myocardin sumoylation transactivates cardiogenic genes in pluripotent 10T1/2 fibroblasts.
Jun Wang,AnKang Li,ZhiGao Wang,XinHua Feng,Eric N Olson,Robert J Schwartz
DOI: https://doi.org/10.1128/MCB.01160-06
IF: 5.069
2007-01-01
Molecular and Cellular Biology
Abstract:Myocardin, a serum response factor (SRF)-dependent cofactor, is a potent activator of smooth muscle gene activity but a poor activator of cardiogenic genes in pluripotent 10T1/2 fibroblasts. Posttranslational modification of GATA4, another myocardin cofactor, by sumoylation strongly activated cardiogenic gene activity. Here, we found that myocardin's activity was strongly enhanced by SUMO-1 via modification of a lysine residue primarily located at position 445 and that the conversion of this residue to arginine (K445R) impaired myocardin transactivation. PIAS1 was involved in governing myocardin activity via its E3 ligase activity that stimulated myocardin sumoylation on an atypical sumoylation site(s) and by its physical association with myocardin. Myocardin initiated the expression of cardiac muscle-specified genes, such as those encoding cardiac a-actin and a-myosin heavy chain, in an SRF-dependent manner in 10T1/2 fibroblasts, but only in the presence of coexpressed SUMO-1/PIAS1. Thus, SUMO modification acted as a molecular switch to promote myocardin's role in cardiogenic gene expression.
-
Mural Cell SRF Controls Pericyte Migration, Vessel Patterning and Blood Flow
Michael M. Orlich,Rodrigo Diéguez-Hurtado,Regine Muehlfriedel,Vithiyanjali Sothilingam,Hartwig Wolburg,Cansu Ebru Oender,Pascal Woelffing,Christer Betsholtz,Konstantin Gaengel,Mathias Seeliger,Ralf H. Adams,Alfred Nordheim
DOI: https://doi.org/10.1161/CIRCRESAHA.122.321109
IF: 23.213
2022-07-16
Circulation Research
Abstract:Pericytes and vascular smooth muscle cells, collectively known as mural cells, are recruited through PDGFB (platelet-derived growth factor B)-PDGFRB (platelet-derived growth factor receptor beta) signaling. MCs are essential for vascular integrity, and their loss has been associated with numerous diseases. Most of this knowledge is based on studies in which MCs are insufficiently recruited or fully absent upon inducible ablation. In contrast, little is known about the physiological consequences that result from impairment of specific MC functions. Here, we characterize the role of the transcription factor SRF (serum response factor) in MCs and study its function in developmental and pathological contexts. We generated a mouse model of MC-specific inducible Srf gene deletion and studied its consequences during retinal angiogenesis using RNA-sequencing, immunohistology, in vivo live imaging, and in vitro techniques. By postnatal day 6, pericytes lacking SRF were morphologically abnormal and failed to properly comigrate with angiogenic sprouts. As a consequence, pericyte-deficient vessels at the retinal sprouting front became dilated and leaky. By postnatal day 12, also the vascular smooth muscle cells had lost SRF, which coincided with the formation of pathological arteriovenous shunts. Mechanistically, we show that PDGFB-dependent SRF activation is mediated via MRTF (myocardin-related transcription factor) cofactors. We further show that MRTF-SRF signaling promotes pathological pericyte activation during ischemic retinopathy. RNA-sequencing, immunohistology, in vivo live imaging, and in vitro experiments demonstrated that SRF regulates expression of contractile SMC proteins essential to maintain the vascular tone. SRF is crucial for distinct functions in pericytes and vascular smooth muscle cells. SRF directs pericyte migration downstream of PDGFRB signaling and mediates pathological pericyte activation during ischemic retinopathy. In vascular smooth muscle cells, SRF is essential for expression of the contractile machinery, and its deletion triggers formation of arteriovenous shunts. These essential roles in physiological and pathological contexts provide a rationale for novel therapeutic approaches through targeting SRF activity in MCs. Download figure Download PowerPoint
cardiac & cardiovascular systems,peripheral vascular disease,hematology
-
Abstract 363: Resident Vascular Adventitial Progenitor Cells Of Smooth Muscle Cell-lineage Adopt Profibrotic Phenotype And Contribute To Cardiac Fibrosis.
Sizhao Lu,Allison Dubner,Austin Jolly,Tysen Noble,Raphael Nemenoff,Karen Moulton,Mary C Weiser-Evans
DOI: https://doi.org/10.1161/atvb.43.suppl_1.363
2023-05-01
Abstract:Cardiac myofibroblasts are the major contributors to ECM deposition in pathological cardiovascular fibrosis, which is the characteristic feature of cardiovascular diseases. However, due to potential heterogeneity of myofibroblasts, the origin of these cells remains controversial. Using highly specific smooth muscle cell lineage-tracing mouse models, we discovered the smooth muscle cell origin of a subpopulation of resident vascular adventitial progenitor cells, defined by the expression of the stem cell marker Sca1 (AdvSca1-SM cells), which rapidly proliferate and adopt a myofibroblast phenotype in response to acute vascular injury. Further, we identified a specific gene signature of active hedgehog/Wnt/β-catenin/Klf4 signaling in AdvSca1-SM cells and validated a Gli1-CreERT2-ROSA26-YFP (Gli1) reporter mouse model to be a faithful lineage tracing system for AdvSca1-SM cells. However, the function of AdvSca1-SM cells in cardiac fibrosis is unknown. Using immunofluorescent staining and label-free second harmonic generation (SHG) imaging, we observed the expansion and migration of AdvSca1-SM cells in close association with perivascular and interstitial cardiac fibrosis in pressure overload-induced cardiac fibrosis in Gli1 reporter mice. By integrating single cell RNA sequencing (scRNA-seq) with spatial transcriptomics, we identified a AngII-driven spatiotemporal profibrotic differentiation trajectory of AdvSca1-SM cells in mouse cardiac tissue. The trajectory, characterized by loss of expression of stemness genes, such as Klf4 , the lncRNA, Meg3 , and up-regulation of myofibroblast genes, was recapitulated in human RNA-seq data and of translational significance. Connectivity map analysis of the scRNA-seq data identified statins as potential candidates for inhibition of the profibrotic transition of AdvSca1-SM cells. In agreement, simvastatin induced the expression of stemness genes and inhibited TGFβ-induced up-regulation of αSMA and down-regulation of KLF4 in cultured AdvSca1-SM cells. Lentiviral-mediated up-regulation of Meg3 also rescued TGFβ-induced suppression of stemness genes. Our findings support the further development of anti-fibrotic therapeutics targeting AdcSca1-SM cells.
peripheral vascular disease,hematology
-
Hierarchical and Stage-Specific Regulation of Murine Cardiomyocyte Maturation by Serum Response Factor.
Yuxuan Guo,Blake D Jardin,Pingzhu Zhou,Isha Sethi,Brynn N Akerberg,Christopher N Toepfer,Yulan Ai,Yifei Li,Qing Ma,Silvia Guatimosim,Yongwu Hu,Grigor Varuzhanyan,Nathan J VanDusen,Donghui Zhang,David C Chan,Guo-Cheng Yuan,Christine E Seidman,Jonathan G Seidman,William T Pu
DOI: https://doi.org/10.1038/s41467-018-06347-2
IF: 16.6
2018-01-01
Nature Communications
Abstract:After birth, cardiomyocytes (CM) acquire numerous adaptations in order to efficiently pump blood throughout an animal's lifespan. How this maturation process is regulated and coordinated is poorly understood. Here, we perform a CRISPR/Cas9 screen in mice and identify serum response factor (SRF) as a key regulator of CM maturation. Mosaic SRF depletion in neonatal CMs disrupts many aspects of their maturation, including sarcomere expansion, mitochondrial biogenesis, transverse-tubule formation, and cellular hypertrophy. Maintenance of maturity in adult CMs is less dependent on SRF. This stage-specific activity is associated with developmentally regulated SRF chromatin occupancy and transcriptional regulation. SRF directly activates genes that regulate sarcomere assembly and mitochondrial dynamics. Perturbation of sarcomere assembly but not mitochondrial dynamics recapitulates SRF knockout phenotypes. SRF overexpression also perturbs CM maturation. Together, these data indicate that carefully balanced SRF activity is essential to promote CM maturation through a hierarchy of cellular processes orchestrated by sarcomere assembly.
-
Interaction of Smad3 and SRF-associated complex mediates TGF-beta1 signals to regulate SM22 transcription during myofibroblast differentiation.
Ping Qiu,Xin Hua Feng,Li Li
DOI: https://doi.org/10.1016/j.yjmcc.2003.09.002
IF: 5.763
2003-01-01
Journal of Molecular and Cellular Cardiology
Abstract:Myofibroblasts play important roles in a variety of developmental and pathological processes, such as vascular remodeling, atherosclerosis and wound healing. In this study, we used the TGF-beta1-treated 10T1/2 cells as an in vitro model to understand how Smad-mediated TGF-beta1 signals regulate SM22 promoter transcription during myofibroblast differentiation. We found that TGF-beta1 transiently induces SRF and SM22 transcription, and that this process is accompanied by transient increases of SRF and Smad3 binding to the SM22 promoter. Interestingly, Smad3, not Smad2, is the primary mediator for TGF-beta1-induced transactivation of the SM22 promoter, while Smad6 and Smad7 repress such a transactivation. Smad3 can bind to a Smad-binding element (SBE) in the first exon of SM22, and directly associate with the SRF complex in response to TGF-beta1 treatment. Moreover, Smad3 and I-Smads regulate the SM22 promoter through CArG box-dependent transcription using dominant-negative SRF mutants and SRF-VP16. Although SBE as well as CArG boxes and TGF-beta control element are all important for the SM22 promoter activities, the promoters with mutations at either one or all of them still respond to TGF-beta1 treatment. Consistently, TGF-beta1 stimulates SM22 transcription in Smad3 null mouse embryonic fibroblasts. These findings provide the first evidence that Smad3 directly links TGF-beta1 signaling to an SRF-associated regulatory network in controlling SM22 transcription; it also implies that TGF-beta1 regulates the SM22 promoter via Smad3-dependent and Smad3-independent pathways.
-
Mutual Potentiation Between Myofibril Assembly and Serum Response Factor in Cardiomyocyte Maturation
Yuxuan Guo,Blake Jardin,Isha Sethi,Behzad Moghadaszadeh,Alan Beggs,William Pu
DOI: https://doi.org/10.1161/res.125.suppl_1.920
IF: 23.213
2019-01-01
Circulation Research
Abstract:Cardiomyocyte (CM) maturation is characterized by transcriptional, morphological and functional specializations that are essential for robust and sustained CM contractions throughout lifetime. The signal networks that govern CM maturation remain poorly defined, which obscures the role of CM maturation in inherited cardiomyopathies and myocardial regeneration and impairs efforts to engineer mature cardiac tissues in vitro . Our prior studies established the transcription factor serum response factor (SRF) as a key regulator of CM maturation: SRF regulates major CM maturation events including myofibril expansion, mitochondria biogenesis, transverse-tubule formation, and cellular hypertrophy. Myofibrillar genes were identified as direct SRF downstream targets that are required for other aspects of CM maturation. To further understand the role of myofibrils in CM maturation, here we report the generation and investigation of a floxed allele of Actn2 in mice, which encodes a central component of sarcomere Z-lines. We applied to these mice a low dose of adeno-associated virus that expressed Cre recombinase specifically in neonatal CMs to generate hearts with mosaic Actn2 mutation. This approach circumvented the confounding secondary effects of cardiac dysfunction in Actn2 mutants and revealed cell-autonomous gene functions. Strikingly, Actn2 ablation triggered dramatic transcriptional dysregulation in addition to the expected myofibrillar disassembly phenotypes in CMs, which strongly correlated with observations in SRF-depleted CMs. Actn2 mutation increased the amount of monomeric actin in CMs, which perturbed the nuclear localization of SRF cofactors MRTF-A/-B. Overexpression of a dominant-negative MRTF-A isoform was sufficient to recapitulate the transcriptional and morphological defects in Actn2 or Srf mutant CMs. Together, these data demonstrate mutual potentiation between myofibril assembly and MRTF-A/B-SRF signaling in CM maturation. This positive feedback loop underlies a novel mechanism by which mechanical forces regulate CM maturation, disruption of which likely contributes to cardiomyopathies caused by sarcomere gene mutations.
-
Crucial role of nrf3 in smooth muscle cell differentiation from stem cells.
Anna Elena Pepe,Qingzhong Xiao,Anna Zampetaki,Zhongyi Zhang,Akira Kobayashi,Yanhua Hu,Qingbo Xu
DOI: https://doi.org/10.1161/CIRCRESAHA.109.211417
IF: 23.213
2010-01-01
Circulation Research
Abstract:Rationale: Nuclear factor erythroid 2-related factor (Nrf)3, a member of the cap 'N' collar family of transcription factors that bind to the DNA-antioxidant responsive elements, is involved in reactive oxygen species balancing and in muscle precursor migration during early embryo development. Objective: To investigate the functional role of Nrf3 in smooth muscle cell (SMC) differentiation in vitro and in vivo. Methods and Results: Nrf3 was upregulated significantly following 1 to 8 days of SMC differentiation. Knockdown of Nrf3 resulted in downregulation of smooth muscle specific markers expression, whereas enforced expression of Nrf3 enhanced SMC differentiation in a dose-dependent manner. SMC-specific transcription factor myocardin, but not serum response factor, was significantly upregulated by Nrf3 overexpression. Strikingly, the binding of SRF and myocardin to the promoter of smooth muscle differentiation genes was dramatically increased by Nrf3 overexpression, and Nrf3 can directly bind to the promoters of SMC differentiation genes as demonstrated by chromatin immunoprecipitation assay. Moreover, NADPH-derived reactive oxygen species production during SMC differentiation was further enhanced by Nrf3 overexpression through upregulation of NADPH oxidase and inhibition of antioxidant signaling pathway. In addition, Nrf3 was involved in the endoplasmic reticulum stressor induced SMC differentiation. Conclusion: Our findings demonstrate for the first time that Nrf3 has a crucial role in SMC differentiation from stem cells indicating that Nrf3 could be a potential target for manipulation of stem cell differentiation toward vascular lineage. (Circ Res. 2010; 106: 870-879.)
-
Abstract 317: Resident Vascular Adventitial Progenitor Cells Of Smooth Muscle Cell-lineage Adopt Profibrotic Phenotype And Contribute To Cardiac Fibrosis
Sizhao Lu,Austin J Jolly,Allison M Dubner,Marie F Mutryn,Raphael A Nemenoff,Karen S Moulton,Mary C Weiser-Evans
DOI: https://doi.org/10.1161/atvb.42.suppl_1.317
2022-05-01
Abstract:Activated cardiac myofibroblasts are the major contributors to ECM deposition in pathological cardiovascular fibrosis, which is the characteristic feature of cardiovascular diseases. However, due to potential heterogeneity of myofibroblasts, the origin of these cells remains controversial. Using highly specific smooth muscle cell lineage-tracing mouse models, we discovered the smooth muscle cell origin of a subpopulation of resident vascular adventitial progenitor cells, defined by the expression of the stem cell marker Sca1 (AdvSca1-SM cells), which rapidly proliferate and adopt a myofibroblast phenotype in response to acute vascular injury. Further, we identified a specific gene signature of active hedgehog/Wnt/β-catenin/ Klf4 signaling in AdvSca1-SM cells and validated a Gli1 -CreERT2-ROSA26-YFP (Gli1) reporter mouse model to be a faithful lineage tracing system for AdvSca1-SM cells. However, the function of AdvSca1-SM cells in cardiac fibrosis is unknown. Using immunofluorescent staining and label-free second harmonic generation (SHG) imaging, we observed the expansion and migration of AdvSca1-SM cells in close association with perivascular and interstitial cardiac fibrosis in pressure overload-induced cardiac fibrosis in Gli1 reporter mice. We performed single cell RNA sequencing (scRNA-seq) to examine the phenotype of AdvSca1-SM cells in Angiotensin II (AngII) model. Our data showed that, upon AngII challenge, AdvSca1-SM cells differentiate along a profibrotic trajectory, which is characterized by loss of expression of Klf4 , the lncRNA, Meg3 , and stemness genes and up-regulation of myofibroblast genes. Importantly, AngII-induced profibrotic transcriptomic changes of AdvSca1-SM cells were recapitulated in human ventricular tissues exhibiting a gene signature of cardiac hypertrophy, emphasizing the translational significance of this phenotypic transition. Connectivity map analysis of the scRNA-seq data identified statins as potential candidates for inhibition of the profibrotic transition of AdvSca1-SM cells. In agreement, simvastatin induced the expression of stemness genes and inhibited TGFβ-induced up-regulation of αSMA and down-regulation of Klf4 in cultured AdvSca1-SM cells.
peripheral vascular disease,hematology
-
Myocardin-Related Transcription Factor A and Yes-Associated Protein Exert Dual Control in G Protein-Coupled Receptor- and RhoA-Mediated Transcriptional Regulation and Cell Proliferation
Olivia M. Yu,Shigeki Miyamoto,Joan Heller Brown
DOI: https://doi.org/10.1128/mcb.00772-15
IF: 5.069
2016-01-01
Molecular and Cellular Biology
Abstract:ABSTRACT The ability of a subset of G protein-coupled receptors (GPCRs) to activate RhoA endows them with unique growth-regulatory properties. Two transcriptional pathways are activated through GPCRs and RhoA, one utilizing the transcriptional coactivator myocardin-related transcription factor A (MRTF-A) and serum response factor (SRF) and the other using the transcriptional coactivator Yes-associated protein (YAP) and TEA domain family members (TEAD). These pathways have not been compared for their relative levels of importance and potential interactions in RhoA target gene expression. GPCRs for thrombin and sphingosine-1-phosphate (S1P) on human glioblastoma cells robustly couple to RhoA and induce the matricelluar protein CCN1. Knockdown of either MRTF-A or YAP abrogates S1P-stimulated CCN1 expression, demonstrating that both coactivators are required. MRTF-A and YAP are also both required for transcriptional control of other S1P-regulated genes in various cell types and for S1P-stimulated glioblastoma cell proliferation. Interactions between MRTF-A and YAP are suggested by their synergistic effects on SRE.L- and TEAD-luciferase expression. Moreover, MRTF-A and YAP associate in coimmunoprecipitations from S1P-stimulated cells. Chromatin immunoprecipitation (ChIP) analysis of the CCN1 gene promoter demonstrated that S1P increases coactivator binding at the canonical transcription factor sequences. Unexpectedly, S1P also enhances MRTF-A binding at TEA sites. Our findings reveal that GPCR- and RhoA-regulated gene expression requires dual input and integration of two distinct transcriptional pathways.
cell biology,biochemistry & molecular biology
-
Abstract 2149: Smooth Muscle-derived Resident Vascular Adventitial Progenitor Cell Contribution To Cardiac Fibrosis And Inflammation Is Dependent On KLF4
Sizhao Lu,Allison M Dubner,Austin J Jolly,Tysen D Noble,Tyler Hinthorn,Raphael A Nemenoff,Karen S Moulton,Mary C Weiser-Evans
DOI: https://doi.org/10.1161/atvb.44.suppl_1.2149
2024-05-01
Abstract:Cardiac fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) material resulting in cardiac tissue scarring and dysfunction. While it is commonly accepted that myofibroblasts are the major contributors to ECM deposition in cardiac fibrosis, their origin remains debated. By combining lineage tracing and RNA sequencing, our group made the paradigm-shifting discovery that a subpopulation of resident vascular stem cells residing within the vascular adventitia (termed AdvSca1-SM cells) originate from mature vascular smooth muscle cells (SMCs) through an in situ reprogramming process. SMC-to-AdvSca1-SM reprogramming and AdvSca1-SM cell maintenance is dependent on induction and activity of the transcription factor, KLF4. In contrast, loss of Klf4 expression promotes the transition of AdvSca1-SM cells to a myofibroblast phenotype in vascular injury and remodeling. However, the molecular mechanism whereby KLF4 regulates AdvSca1-SM phenotype remains unclear. Our recent preliminary data confirm the existence of perivascular cardiac AdvSca1-SM cells. Leveraging a highly specific AdvSca1-SM cell reporter system, single-cell RNA-sequencing (scRNA-seq), and spatial transcriptomic approaches, we demonstrate the profibrotic differentiation trajectory of AdvSca1-SM cells in the setting of Angiotensin II (AngII)-induced cardiac fibrosis. Differentiation is characterized by loss of stemness-related genes, including Klf4 , but gain of expression of a profibrotic phenotype. Importantly, these changes are recapitulated in human cardiac hypertrophic tissue, supporting the translational significance of profibrotic transition of AdvSca1-SM-like cells in human cardiomyopathy. Very surprisingly and paradoxically, AdvSca1-SM-specific genetic knockout of Klf4 prior to AngII treatment protects against cardiac inflammation and fibrosis, indicating that Klf4 is essential for the profibrotic response of AdvSca1-SM cells. Overall, our data reveal the contribution of AdvSca1-SM cells to myofibroblasts in the setting of cardiac fibrosis. KLF4 not only maintains the stemness of AdvSca1-SM cells, but also orchestrates their response to profibrotic stimuli, and may serve as a therapeutic target in cardiac fibrosis.
peripheral vascular disease,hematology