1
|
Bruijn LE, van den Akker BEWM, van Rhijn CM, Hamming JF, Lindeman JHN. Extreme Diversity of the Human Vascular Mesenchymal Cell Landscape. J Am Heart Assoc 2020; 9:e017094. [PMID: 33190596 PMCID: PMC7763765 DOI: 10.1161/jaha.120.017094] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Background Human mesenchymal cells are culprit factors in vascular (patho)physiology and are hallmarked by phenotypic and functional heterogeneity. At present, they are subdivided by classic umbrella terms, such as "fibroblasts," "myofibroblasts," "smooth muscle cells," "fibrocytes," "mesangial cells," and "pericytes." However, a discriminative marker-based subclassification has to date not been established. Methods and Results As a first effort toward a classification scheme, a systematic literature search was performed to identify the most commonly used phenotypical and functional protein markers for characterizing and classifying vascular mesenchymal cell subpopulation(s). We next applied immunohistochemistry and immunofluorescence to inventory the expression pattern of identified markers on human aorta specimens representing early, intermediate, and end stages of human atherosclerotic disease. Included markers comprise markers for mesenchymal lineage (vimentin, FSP-1 [fibroblast-specific protein-1]/S100A4, cluster of differentiation (CD) 90/thymocyte differentiation antigen 1, and FAP [fibroblast activation protein]), contractile/non-contractile phenotype (α-smooth muscle actin, smooth muscle myosin heavy chain, and nonmuscle myosin heavy chain), and auxiliary contractile markers (h1-Calponin, h-Caldesmon, Desmin, SM22α [smooth muscle protein 22α], non-muscle myosin heavy chain, smooth muscle myosin heavy chain, Smoothelin-B, α-Tropomyosin, and Telokin) or adhesion proteins (Paxillin and Vinculin). Vimentin classified as the most inclusive lineage marker. Subset markers did not separate along classic lines of smooth muscle cell, myofibroblast, or fibroblast, but showed clear temporal and spatial diversity. Strong indications were found for presence of stem cells/Endothelial-to-Mesenchymal cell Transition and fibrocytes in specific aspects of the human atherosclerotic process. Conclusions This systematic evaluation shows a highly diverse and dynamic landscape for the human vascular mesenchymal cell population that is not captured by the classic nomenclature. Our observations stress the need for a consensus multiparameter subclass designation along the lines of the cluster of differentiation classification for leucocytes.
Collapse
Affiliation(s)
- Laura E. Bruijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | | | - Connie M. van Rhijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jaap F. Hamming
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jan H. N. Lindeman
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
2
|
Gu W, Hong X, Potter C, Qu A, Xu Q. Mesenchymal stem cells and vascular regeneration. Microcirculation 2018; 24. [PMID: 27681821 DOI: 10.1111/micc.12324] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022]
Abstract
In recent years, MSCs have emerged as a promising therapeutic cell type in regenerative medicine. They hold great promise for treating cardiovascular diseases, such as myocardial infarction and limb ischemia. MSCs may be utilized in both cell-based therapy and vascular graft engineering to restore vascular function, thereby providing therapeutic benefits to patients. The efficacy of MSCs lies in their multipotent differentiation ability toward vascular smooth muscle cells, endothelial cells and other cell types, as well as their capacity to secrete various trophic factors, which are potent in promoting angiogenesis, inhibiting apoptosis and modulating immunoreaction. Increasing our understanding of the mechanisms of MSC involvement in vascular regeneration will be beneficial in boosting present therapeutic approaches and developing novel ones to treat cardiovascular diseases. In this review, we aim to summarize current progress in characterizing the in vivo identity of MSCs, to discuss mechanisms involved in cell-based therapy utilizing MSCs, and to explore current and future strategies for vascular regeneration.
Collapse
Affiliation(s)
- Wenduo Gu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Xuechong Hong
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Claire Potter
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
3
|
Starke RM, Thompson JW, Ali MS, Pascale CL, Martinez Lege A, Ding D, Chalouhi N, Hasan DM, Jabbour P, Owens GK, Toborek M, Hare JM, Dumont AS. Cigarette Smoke Initiates Oxidative Stress-Induced Cellular Phenotypic Modulation Leading to Cerebral Aneurysm Pathogenesis. Arterioscler Thromb Vasc Biol 2018; 38:610-621. [PMID: 29348119 DOI: 10.1161/atvbaha.117.310478] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Cigarette smoke exposure (CSE) is a risk factor for cerebral aneurysm (CA) formation, but the molecular mechanisms are unclear. Although CSE is known to contribute to excess reactive oxygen species generation, the role of oxidative stress on vascular smooth muscle cell (VSMC) phenotypic modulation and pathogenesis of CAs is unknown. The goal of this study was to investigate whether CSE activates a NOX (NADPH oxidase)-dependent pathway leading to VSMC phenotypic modulation and CA formation and rupture. APPROACH AND RESULTS In cultured cerebral VSMCs, CSE increased expression of NOX1 and reactive oxygen species which preceded upregulation of proinflammatory/matrix remodeling genes (MCP-1, MMPs [matrix metalloproteinase], TNF-α, IL-1β, NF-κB, KLF4 [Kruppel-like factor 4]) and downregulation of contractile genes (SM-α-actin [smooth muscle α actin], SM-22α [smooth muscle 22α], SM-MHC [smooth muscle myosin heavy chain]) and myocardin. Inhibition of reactive oxygen species production and knockdown of NOX1 with siRNA or antisense decreased CSE-induced upregulation of NOX1 and inflammatory genes and downregulation of VSMC contractile genes and myocardin. p47phox-/- NOX knockout mice, or pretreatment with the NOX inhibitor, apocynin, significantly decreased CA formation and rupture compared with controls. NOX1 protein and mRNA expression were similar in p47phox-/- mice and those pretreated with apocynin but were elevated in unruptured and ruptured CAs. CSE increased CA formation and rupture, which was diminished with apocynin pretreatment. Similarly, NOX1 protein and mRNA and reactive oxygen species were elevated by CSE, and in unruptured and ruptured CAs. CONCLUSIONS CSE initiates oxidative stress-induced phenotypic modulation of VSMCs and CA formation and rupture. These molecular changes implicate oxidative stress in the pathogenesis of CAs and may provide a potential target for future therapeutic strategies.
Collapse
Affiliation(s)
- Robert M Starke
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.).
| | - John W Thompson
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Muhammad S Ali
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Crissey L Pascale
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Alejandra Martinez Lege
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Dale Ding
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Nohra Chalouhi
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - David M Hasan
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Pascal Jabbour
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Gary K Owens
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Michal Toborek
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Joshua M Hare
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| | - Aaron S Dumont
- From the Department of Neurological Surgery & Radiology, University of Miami Cerebrovascular Initiative (R.M.S., J.W.T.), Department of Biochemistry and Molecular Biology (M.T.), and Department of Cardiology and Molecular and Cellular Pharmacology (J.M.H.), University of Miami, FL; Department of Neurosurgery, University of Iowa, Iowa City (M.S.A., D.M.H.); Department of Neurological Surgery, Tulane University, New Orleans, LA (C.L.P., A.M.L., A.S.D.); Department of Neurosurgery (D.D.) and Department of Molecular Physiology & Biophysics, Robert M. Berne Cardiovascular Research Center (G.K.O.), University of Virginia, Charlottesville; and Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA (N.C., P.J.)
| |
Collapse
|
4
|
Tang Y, Yu S, Liu Y, Zhang J, Han L, Xu Z. MicroRNA-124 controls human vascular smooth muscle cell phenotypic switch via Sp1. Am J Physiol Heart Circ Physiol 2017; 313:H641-H649. [PMID: 28667053 DOI: 10.1152/ajpheart.00660.2016] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 06/26/2017] [Accepted: 06/26/2017] [Indexed: 01/07/2023]
Abstract
Phenotypic switch of vascular smooth muscle cells (VSMCs) plays an important role in the pathogenesis of atherosclerosis and aortic dissection. However, the mechanisms of phenotypic modulation are still unclear. MicroRNAs have emerged as important regulators of VSMC function. We recently found that microRNA-124 (miR-124) was downregulated in proliferative vascular diseases that were characterized by a VSMC phenotypic switch. Therefore, we speculated that the aberrant expression of miR-124 might play a critical role in human aortic VSMC phenotypic switch. Using quantitative RT-PCR, we found that miR-124 was dramatically downregulated in the aortic media of clinical specimens of the dissected aorta and correlated with molecular markers of the contractile VSMC phenotype. Overexpression of miR-124 by mimicking transfection significantly attenuated platelet-derived growth factor-BB-induced human aortic VSMC proliferation and phenotypic switch. Furthermore, we identified specificity protein 1 (Sp1) as the downstream target of miR-124. A luciferase reporter assay was used to confirm direct miR-124 targeting of the 3'-untranslated region of the Sp1 gene and repression of Sp1 expression in human aortic VSMCs. Furthermore, constitutively active Sp1 in miR-124-overexpressing VSMCs reversed the antiproliferative effects of miR-124. These results demonstrated a novel mechanism of miR-124 modulation of VSMC phenotypic switch by targeting Sp1 expression.NEW & NOTEWORTHY Previous studies have demonstrated that miR-124 is involved in the proliferation of a variety of cell types. However, miRNAs are expressed in a tissue-specific manner. We first identified miR-124 as a critical regulator in human aortic vascular smooth muscle cell differentiation, proliferation, and phenotype switch by targeting the 3'-untranslated region of specificity protein 1.
Collapse
Affiliation(s)
- Yangfeng Tang
- Changhai Hospital of the Second Military College, Shanghai, China
| | - Shangyi Yu
- Changhai Hospital of the Second Military College, Shanghai, China
| | - Yang Liu
- Changhai Hospital of the Second Military College, Shanghai, China
| | - Jiajun Zhang
- Changhai Hospital of the Second Military College, Shanghai, China
| | - Lin Han
- Changhai Hospital of the Second Military College, Shanghai, China
| | - Zhiyun Xu
- Changhai Hospital of the Second Military College, Shanghai, China
| |
Collapse
|
5
|
Seruya M, Shah A, Pedrotty D, du Laney T, Melgiri R, McKee JA, Young HE, Niklason LE. Clonal Population of Adult Stem Cells: Life Span and Differentiation Potential. Cell Transplant 2017; 13:93-101. [PMID: 15129755 DOI: 10.3727/000000004773301762] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Adult stem cells derived from bone marrow, connective tissue, and solid organs can exhibit a range of differentiation potentials. Some controversy exists regarding the classification of mesenchymal stem cells as bona fide stem cells, which is in part derived from the limited ability to propagate true clonal populations of precursor cells. We isolated putative mesenchymal stem cells from the connective tissue of an adult rat (rMSC), and generated clonal populations via three rounds of dilutional cloning. The replicative potential of the clonal rMSC line far exceeded Hayflick's limit of 50–70 population doublings. The high capacity for self-renewal in vitro correlated with telomerase activity, as demonstrated by telomerase repeat amplification protocol (TRAP) assay. Exposure to nonspecific differentiation culture medium revealed multilineage differentiation potential of rMSC clones. Immunostaining confirmed the appearance of mesodermal phenotypes, including adipocytes possessing lipid-rich vacuoles, chondrocytes depositing pericellular type II collagen, and skeletal myoblasts expressing MyoD1. Importantly, the spectrum of differentiation capability was sustained through repeated passaging. Furthermore, serum-free conditions that led to high-efficiency smooth muscle differentiation were identified. rMSCs plated on collagen IV-coated surfaces and exposed to transforming growth factor-β1 (TGF-β1) differentiated into a homogeneous population expressing α-actin and calponin. Hence, clonogenic analysis confirmed the presence of a putative MSC population derived from the connective tissue of rat skeletal muscle. The ability to differentiate into a smooth muscle cell (SMC) phenotype, combined with a high proliferative capacity, make such a connective tissue-derived MSC population ideal for applications in vascular tissue construction.
Collapse
Affiliation(s)
- Mitchel Seruya
- Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Regulation of actin dynamics by WNT-5A: implications for human airway smooth muscle contraction. Sci Rep 2016; 6:30676. [PMID: 27468699 PMCID: PMC4965744 DOI: 10.1038/srep30676] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 07/07/2016] [Indexed: 01/13/2023] Open
Abstract
A defining feature of asthma is airway hyperresponsiveness (AHR), which underlies the exaggerated bronchoconstriction response of asthmatics. The role of the airway smooth muscle (ASM) in AHR has garnered increasing interest over the years, but how asthmatic ASM differs from healthy ASM is still an active topic of debate. WNT-5A is increasingly expressed in asthmatic ASM and has been linked with Th2-high asthma. Due to its link with calcium and cytoskeletal remodelling, we propose that WNT-5A may modulate ASM contractility. We demonstrated that WNT-5A can increase maximum isometric tension in bovine tracheal smooth muscle strips. In addition, we show that WNT-5A is preferentially expressed in contractile human airway myocytes compared to proliferative cells, suggesting an active role in maintaining contractility. Furthermore, WNT-5A treatment drives actin polymerisation, but has no effect on intracellular calcium flux. Next, we demonstrated that WNT-5A directly regulates TGF-β1-induced expression of α-SMA via ROCK-mediated actin polymerization. These findings suggest that WNT-5A modulates fundamental mechanisms that affect ASM contraction and thus may be of relevance for AHR in asthma.
Collapse
|
7
|
Rozenberg JM, Tesfu DB, Musunuri S, Taylor JM, Mack CP. DNA methylation of a GC repressor element in the smooth muscle myosin heavy chain promoter facilitates binding of the Notch-associated transcription factor, RBPJ/CSL1. Arterioscler Thromb Vasc Biol 2014; 34:2624-31. [PMID: 25324571 DOI: 10.1161/atvbaha.114.304634] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The goal of the present study was to identify novel mechanisms that regulate smooth muscle cell (SMC) differentiation marker gene expression. APPROACH AND RESULTS We demonstrate that the CArG-containing regions of many SMC-specific promoters are imbedded within CpG islands. A previously identified GC repressor element in the SM myosin heavy chain (MHC) promoter was highly methylated in cultured aortic SMC but not in the aorta, and this difference was inversely correlated with SM MHC expression. Using an affinity chromatography/mass spectroscopy-based approach, we identified the multifunctional Notch transcription factor, recombination signal binding protein for immunoglobulin κ J region (RBPJ), as a methylated GC repressor-binding protein. RBPJ protein levels and binding to the endogenous SM MHC GC repressor were enhanced by platelet-derived growth factor-BB treatment. A methylation mimetic mutation to the GC repressor that facilitated RBPJ binding inhibited SM MHC promoter activity as did overexpression of RBPJ. Consistent with this, knockdown of RBPJ in phenotypically modulated human aortic SMC enhanced endogenous SMC marker gene expression, an effect likely mediated by increased recruitment of serum response factor and Pol II to the SMC-specific promoters. In contrast, the depletion of RBPJ in differentiated transforming growth factor-β-treated SMC inhibited SMC-specific gene activation, supporting the idea that the effects of RBPJ/Notch signaling are context dependent. CONCLUSIONS Our results indicate that methylation-dependent binding of RBPJ to a GC repressor element can negatively regulate SM MHC promoter activity and that RBPJ can inhibit SMC marker gene expression in phenotypically modulated SMC. These results will have important implications on the regulation of SMC phenotype and on Notch-dependent transcription.
Collapse
Affiliation(s)
- Julian M Rozenberg
- From the Department of Pathology, University of North Carolina, Chapel Hill
| | - Daniel B Tesfu
- From the Department of Pathology, University of North Carolina, Chapel Hill
| | - Srilaxmi Musunuri
- From the Department of Pathology, University of North Carolina, Chapel Hill
| | - Joan M Taylor
- From the Department of Pathology, University of North Carolina, Chapel Hill
| | - Christopher P Mack
- From the Department of Pathology, University of North Carolina, Chapel Hill.
| |
Collapse
|
8
|
YU XIN, LI ZHENG. MicroRNAs regulate vascular smooth muscle cell functions in atherosclerosis (Review). Int J Mol Med 2014; 34:923-33. [DOI: 10.3892/ijmm.2014.1853] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 05/30/2014] [Indexed: 11/06/2022] Open
|
9
|
Starke RM, Ali MS, Jabbour PM, Tjoumakaris SI, Gonzalez F, Hasan DM, Rosenwasser RH, Owens GK, Koch WJ, Dumont AS. Cigarette smoke modulates vascular smooth muscle phenotype: implications for carotid and cerebrovascular disease. PLoS One 2013; 8:e71954. [PMID: 23967268 PMCID: PMC3743809 DOI: 10.1371/journal.pone.0071954] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 07/05/2013] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The role of smooth muscle cell (SMC) phenotypic modulation in the cerebral circulation and pathogenesis of stroke has not been determined. Cigarette smoke is a major risk factor for atherosclerosis, but potential mechanisms are unclear, and its role in SMC phenotypic modulation has not been established. METHODS AND RESULTS In cultured cerebral vascular SMCs, exposure to cigarette smoke extract (CSE) resulted in decreased promoter activity and mRNA expression of key SMC contractile genes (SM-α-actin, SM-22α, SM-MHC) and the transcription factor myocardin in a dose-dependent manner. CSE also induced pro-inflammatory/matrix remodeling genes (MCP-1, MMPs, TNF-α, IL-1β, NF-κB). CSE increased expression of KLF4, a known regulator of SMC differentiation, and siKLF4 inhibited CSE induced suppression of SMC contractile genes and myocardin and activation of inflammatory genes. These mechanisms were confirmed in vivo following exposure of rat carotid arteries to CSE. Chromatin immune-precipitation assays in vivo and in vitro demonstrated that CSE promotes epigenetic changes with binding of KLF4 to the promoter regions of myocardin and SMC marker genes and alterations in promoter acetylation and methylation. CONCLUSION CSE exposure results in phenotypic modulation of cerebral SMC through myocardin and KLF4 dependent mechanisms. These results provides a mechanism by which cigarette smoke induces a pro-inflammatory/matrix remodeling phenotype in SMC and an important pathway for cigarette smoke to contribute to atherosclerosis and stroke.
Collapse
MESH Headings
- Acetylation/drug effects
- Animals
- Carotid Arteries/cytology
- Carotid Arteries/drug effects
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Cell Differentiation/drug effects
- Cerebrovascular Disorders/chemically induced
- Cerebrovascular Disorders/genetics
- Cerebrovascular Disorders/pathology
- DNA Methylation/drug effects
- Down-Regulation/drug effects
- Genetic Markers/genetics
- Histone Deacetylase 2/metabolism
- Histones/metabolism
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/antagonists & inhibitors
- Kruppel-Like Transcription Factors/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Nuclear Proteins/genetics
- Phenotype
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- Rats
- Rats, Sprague-Dawley
- Smoke/adverse effects
- Tobacco Products/analysis
- Trans-Activators/genetics
Collapse
Affiliation(s)
- Robert M. Starke
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Muhammad S. Ali
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Pascal M. Jabbour
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Stavropoula I. Tjoumakaris
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Fernando Gonzalez
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - David M. Hasan
- Department of Neurosurgery, University of Iowa, Iowa City, Iowa, United States of America
| | - Robert H. Rosenwasser
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Gary K. Owens
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, Charlottesville, Virginia, United States of America
| | - Walter J. Koch
- Center for Translational Medicine and Department of Pharmacology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Aaron S. Dumont
- Joseph and Marie Field Cerebrovascular Research Laboratory, Division of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
10
|
Chen Y, Wong MM, Campagnolo P, Simpson R, Winkler B, Margariti A, Hu Y, Xu Q. Adventitial stem cells in vein grafts display multilineage potential that contributes to neointimal formation. Arterioscler Thromb Vasc Biol 2013; 33:1844-51. [PMID: 23744989 DOI: 10.1161/atvbaha.113.300902] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This study was designed to carry out the characterization of stem cells within the adventitia and to elucidate their functional role in the pathogenesis of vein graft atherosclerosis. APPROACH AND RESULTS A mouse vein graft model was used to investigate the functional role of adventitial stem/progenitor cells on atherosclerosis. The adventitia of vein grafts underwent significant remodeling during early stages of vessel grafting and displayed markedly heterogeneous cell compositions. Immunofluorescence staining indicated a significant number of stem cell antigen-1-positive cells that were closely located to vasa vasorum. In vitro clonogenic assays demonstrated 1% to 11% of growing rates from adventitial cell cultures, most of which could be differentiated into smooth muscle cells (SMCs). These stem cell antigen-1-positive cells also displayed a potential to differentiate into adipogenic, osteogenic, or chondrogenic lineages in vitro. In light of the proatherogenic roles of SMCs in atherosclerosis, we focused on the functional roles of progenitor-SMC differentiation, in which we subsequently demonstrated that it was driven by direct interaction of the integrin/collagen IV axis. The ex vivo bioreactor system revealed the migratory capacity of stem cell antigen-1-positive progenitor cells into the vessel wall in response to stromal cell-derived factor-1. Stem cell antigen-1-positive cells that were applied to the outer layer of vein grafts showed enhanced atherosclerosis in apolipoprotein E-deficient mice, which contributed to ≈ 30% of neointimal SMCs. CONCLUSIONS We demonstrate that during pathological conditions in vein grafting, the adventitia harbors stem/progenitor cells that can actively participate in the pathogenesis of vascular disease via differentiation into SMCs.
Collapse
Affiliation(s)
- Yikuan Chen
- Department of Vascular Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Lee MY, Garvey SM, Ripley ML, Wamhoff BR. Genome-wide microarray analyses identify the protein C receptor as a novel calcineurin/nuclear factor of activated T cells-dependent gene in vascular smooth muscle cell phenotypic modulation. Arterioscler Thromb Vasc Biol 2012; 31:2665-75. [PMID: 21903947 DOI: 10.1161/atvbaha.111.235960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Calcineurin (Cn) and the nuclear factor of activated T cells (NFAT) family of transcription factors are critical in vascular smooth muscle cell (SMC) development and pathology. Here, we used a genomics approach to identify and validate NFAT gene targets activated during platelet-derived growth factor-BB (PDGF-BB)-induced SMC phenotypic modulation. METHODS AND RESULTS Genome-wide expression arrays were used to identify genes both (1) differentially activated in response to PDGF-BB and (2) whose differential expression was reduced by both the Cn inhibitor cyclosporin A and the NFAT inhibitor A-285222. The 20 most pharmacologically sensitive genes were validated by quantitative reverse transcription-polymerase chain reaction analysis of PDGF-BB-stimulated SMCs in the presence of Cn/NFAT inhibitors, including the VIVIT peptide. In all experiments, protein C receptor (PROCR) gene activation was reduced. We showed that PROCR expression was virtually absent in untreated, quiescent SMCs. PDGF-BB stimulation, however, induced significant PROCR promoter activation and downstream protein expression in a Cn/NFAT-dependent manner. Mutation of a species-conserved, NFAT binding motif significantly attenuated PDGF-BB-induced PROCR promoter activity, thereby distinguishing NFAT as the first PROCR transcriptional activator to date. Moreover, SMC PROCR expression was upregulated in the neointima as early as 7 days following acute vascular injury in rat carotid arteries. CONCLUSION We hereby report PROCR as a novel, NFAT-dependent gene that may be implicated in vascular restenosis and consequent inward remodeling.
Collapse
Affiliation(s)
- Monica Y Lee
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
12
|
Torella D, Iaconetti C, Catalucci D, Ellison GM, Leone A, Waring CD, Bochicchio A, Vicinanza C, Aquila I, Curcio A, Condorelli G, Indolfi C. MicroRNA-133 Controls Vascular Smooth Muscle Cell Phenotypic Switch In Vitro and Vascular Remodeling In Vivo. Circ Res 2011; 109:880-93. [DOI: 10.1161/circresaha.111.240150] [Citation(s) in RCA: 239] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Daniele Torella
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Claudio Iaconetti
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Daniele Catalucci
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Georgina M. Ellison
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Angelo Leone
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Cheryl D. Waring
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Angela Bochicchio
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Carla Vicinanza
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Iolanda Aquila
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Antonio Curcio
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Gianluigi Condorelli
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| | - Ciro Indolfi
- From the Laboratory of Molecular and Cellular Cardiology, Cardiovascular Institute (D.T., C. Iaconetti, G.M.E., A.L., A.B., C.V., I.A., A.C., C. Indolfi), Magna Graecia University, Catanzaro, Italy; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University (D.T., G.M.E., C.D.W.), Liverpool, United Kingdom; Istituto di Ricovero e Cura a Carattere Scientifico Multimedica (D.C., G.C.), Milan, Italy; (G.C.), Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle
| |
Collapse
|
13
|
Park C, Hennig GW, Sanders KM, Cho JH, Hatton WJ, Redelman D, Park JK, Ward SM, Miano JM, Yan W, Ro S. Serum response factor-dependent MicroRNAs regulate gastrointestinal smooth muscle cell phenotypes. Gastroenterology 2011; 141:164-175. [PMID: 21473868 PMCID: PMC3129374 DOI: 10.1053/j.gastro.2011.03.058] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 02/15/2011] [Accepted: 03/18/2011] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Smooth muscle cells (SMCs) change phenotypes under various pathophysiological conditions. These changes are largely controlled by the serum response factor (SRF), a transcription factor that binds to CC (A/T)6 GG (CArG) boxes in SM contractile genes. MicroRNAs (miRNA) regulate transitions among SMC phenotypes. The SMC miRNA transcriptome (SMC miRNAome) and its regulation by SRF have not been determined. METHODS We performed massively parallel sequencing to identify gastrointestinal (GI) SMC miRNA transcriptomes in mice and humans. SMC miRNA transcriptomes were mapped to identify all CArG boxes, which were confirmed by SRF knockdown and microarrays. Quantitative polymerase chain reaction was used to identify SMC-phenotypic miRNAs in differentiated and proliferating SMCs. Bioinformatics and target validation analysis showed regulation of SMC phenotype by SRF-dependent, SMC-phenotype miRNAs. RESULTS We cloned and identified GI miRNA transcriptomes using genome-wide analyses of mouse and human cells. The SM miRNAome consisted of hundreds of unique miRNAs that were highly conserved among both species. We mapped miRNAs CArG boxes and found that many had an SRF-dependent signature in the SM miRNAome. The SM miRNAs CArG boxes had several distinct features. We also identified approximately 100 SMC-phenotypic miRNAs that were induced in differentiated or proliferative SMC phenotypes. We showed that SRF-dependent, SMC-phenotypic miRNAs bind and regulate Srf and its cofactors, myocadin (Myocd) and member of ETS oncogene family Elk1. CONCLUSIONS The GI SMC phenotypes are controlled by SRF-dependent, SMC-phenotypic miRNAs that regulate expression of SRF, MYOCD, and ELK1.
Collapse
Affiliation(s)
- Chanjae Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| | - Grant W. Hennig
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| | - Jonathan H. Cho
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| | - William J. Hatton
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| | - Doug Redelman
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| | - Jong Kun Park
- Division of Biological Science, Wonkwang University, Iksan, Chonbuk, South Korea
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| | - Joseph M. Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, United States of America
| |
Collapse
|
14
|
Culture media for the differentiation of mesenchymal stromal cells. Acta Biomater 2011; 7:463-77. [PMID: 20688199 DOI: 10.1016/j.actbio.2010.07.037] [Citation(s) in RCA: 192] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 07/20/2010] [Accepted: 07/27/2010] [Indexed: 02/08/2023]
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues such as bone marrow aspirates, fat or umbilical cord blood. These cells have the ability to proliferate in vitro and differentiate into a series of mesoderm-type lineages, including osteoblasts, chondrocytes, adipocytes, myocytes and vascular cells. Due to this ability, MSCs provide an appealing source of progenitor cells which may be used in the field of tissue regeneration for both research and clinical purposes. The key factors for successful MSC proliferation and differentiation in vitro are the culture conditions. Hence, we here summarize the culture media and their compositions currently available for the differentiation of MSCs towards osteogenic, chondrogenic, adipogenic, endothelial and vascular smooth muscle phenotypes. However, optimal combination of growth factors, cytokines and serum supplements and their concentration within the media is essential for the in vitro culture and differentiation of MSCs and thereby for their application in advanced tissue engineering.
Collapse
|
15
|
Weber AA, Schrör K. The significance of platelet-derived growth factors for proliferation of vascular smooth muscle cells. Platelets 2010. [DOI: 10.1080/09537109909169169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
16
|
Gong Z, Calkins G, Cheng EC, Krause D, Niklason LE. Influence of culture medium on smooth muscle cell differentiation from human bone marrow-derived mesenchymal stem cells. Tissue Eng Part A 2009; 15:319-30. [PMID: 19115826 DOI: 10.1089/ten.tea.2008.0161] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (hMSCs) represent an appealing source of smooth muscle cells (SMCs) for engineering small-diameter vascular grafts due to the limited availability and replicative capacity of somatic SMCs. However, lack of standardization of hMSC culture conditions has limited some progress in hMSC research. Because, at the moment, a chemically defined, serum-free medium without growth factors is not capable of amplifying hMSCs in vitro, the usage of serum (either human serum or fetal bovine serum [FBS]) continues in hMSC research. The emergence of commercial hMSCs and hMSC media opened a series of questions regarding the compatibility of commercial and homemade hMSCs and hMSC media. In this study, two types of commonly used FBS-containing hMSC media-MSCGM (containing 10% FBS) and MesenPro (containing 2% FBS), along with our homemade medium (low-glucose Dulbecco's modified Eagle's medium plus 10% selected lot FBS)-were compared in their ability to support SMC differentiation from hMSCs. The effects of FBS level, medium supplements (ascorbic acid, copper, etc.), and growth factors (transforming growth factor beta1) were also examined for their impact on SMC differentiation. It was discovered that MesenPro and transforming growth factor beta1 are the strongest SMC inducers from hMSCs. In contrast, hMSCs grown in homemade (10% Dulbecco's modified Eagle's medium) and commercial MSCGM media remained undifferentiated. FBS concentration did not affect SMC differentiation when 10% FBS was compared with 2%. Finally, the mechanism underlying SMC differentiation from hMSCs grown in FBS-containing medium was explored by following the expression changes of serum response factor during the establishment of hMSC culture.
Collapse
Affiliation(s)
- Zhaodi Gong
- Department of Anesthesiology & Biomedical Engineering, Vascular Biology & Transplantation (VBT) Program, Yale University School of Medicine, New Haven, Connecticut 06520-8089, USA
| | | | | | | | | |
Collapse
|
17
|
Blaker AL, Taylor JM, Mack CP. PKA-dependent phosphorylation of serum response factor inhibits smooth muscle-specific gene expression. Arterioscler Thromb Vasc Biol 2009; 29:2153-60. [PMID: 19778940 DOI: 10.1161/atvbaha.109.197285] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Our goal was to identify phosphorylation sites that regulate serum response factor (SRF) activity to gain a better understanding of the signaling mechanisms that regulate SRF's involvement in smooth muscle cell (SMC)-specific and early response gene expression. METHODS AND RESULTS By screening phosphorylation-deficient and mimetic mutations in SRF(-/-) embryonic stem cells, we identified T159 as a phosphorylation site that significantly inhibits SMC-specific gene expression in an embryonic stem cell model of SMC differentiation. This residue conforms to a highly conserved consensus cAMP-dependent protein kinase (PKA) site, and in vitro and in vivo labeling studies demonstrated that it was phosphorylated by PKA. Results from gel shift and chromatin immunoprecipitation assays demonstrated that T159 phosphorylation inhibited SRF binding to SMC-specific CArG elements. Interestingly, the myocardin factors could at least partially rescue the effects of the T159D mutation under some conditions, but this response was promoter specific. Finally, PKA signaling had much less of an effect on c-fos promoter activity and SRF binding to the c-fos CArG. CONCLUSIONS Our results indicate that phosphorylation of SRF by PKA inhibits SMC-specific transcription suggesting a novel signaling mechanism for the control of SMC phenotype.
Collapse
Affiliation(s)
- Alicia L Blaker
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599-7525, USA
| | | | | |
Collapse
|
18
|
Deaton RA, Gan Q, Owens GK. Sp1-dependent activation of KLF4 is required for PDGF-BB-induced phenotypic modulation of smooth muscle. Am J Physiol Heart Circ Physiol 2009; 296:H1027-37. [PMID: 19168719 DOI: 10.1152/ajpheart.01230.2008] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is clear evidence that the phenotypic modulation of smooth muscle cells (SMCs) contributes to the pathophysiology of vascular disease. Phenotypic modulation refers to the unique ability of SMCs to alter their phenotype in response to extracellular stimuli and is hallmarked by the loss of SMC marker gene expression. The transcription factor Krüppel-like factor 4 (KLF4) is a known powerful negative regulator of SMC marker gene expression that works, in part, by decreasing the expression of the serum response factor (SRF) myocardin. KLF4 is not expressed in healthy adult SMCs but is increased in SMCs in response to vascular injury in vivo or PDGF-BB treatment in vitro. The aim of the present study was to determine the molecular mechanisms that regulate the expression of KLF4 in phenotypically modulated SMCs. The results demonstrated that the transcription factor stimulating protein-1 (Sp1) regulated the expression of KLF4 in SMCs. The KLF4 promoter contains three consensus Sp1 binding sites. Using a series of truncated KLF4 promoters, we showed that only fragments containing these Sp1 sites could be activated by PDGF-BB. In addition, overexpression of Sp1 alone was sufficient to increase the activity of the KLF4 promoter. Moreover, inhibiting Sp1 expression with small-interfering RNA attenuated the effects of PDGF-BB on KLF4 expression. Mutation of the three Sp1 sites within the KLF4 promoter abolished both baseline and PDGF-BB-induced activity. Finally, the results demonstrated enhanced Sp1 binding to the KLF4 promoter in SMCs treated with PDGF-BB in vitro and following vascular injury in vivo. Taken together, the results suggest a novel role for Sp1 in increasing the expression of KLF4 in phenotypically modulated SMCs.
Collapse
Affiliation(s)
- Rebecca A Deaton
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | | | | |
Collapse
|
19
|
Statins activate GATA-6 and induce differentiated vascular smooth muscle cells. Biochem Biophys Res Commun 2008; 374:731-6. [DOI: 10.1016/j.bbrc.2008.07.098] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Accepted: 07/18/2008] [Indexed: 11/20/2022]
|
20
|
Petit MM, Lindskog H, Larsson E, Wasteson P, Athley E, Breuer S, Angstenberger M, Hertfelder D, Mattsson E, Nordheim A, Nelander S, Lindahl P. Smooth Muscle Expression of Lipoma Preferred Partner Is Mediated by an Alternative Intronic Promoter That Is Regulated by Serum Response Factor/Myocardin. Circ Res 2008; 103:61-9. [DOI: 10.1161/circresaha.108.177436] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipoma preferred partner (LPP) was recently recognized as a smooth muscle marker that plays a role in smooth muscle cell migration. In this report, we focus on the transcriptional regulation of the LPP gene. In particular, we investigate whether LPP is directly regulated by serum response factor (SRF). We show that the LPP gene contains 3 evolutionarily conserved CArG boxes and that 1 of these is part of an alternative promoter in intron 2. Quantitative RT-PCR shows that this alternative promoter directs transcription specifically to smooth muscle containing tissues in vivo. By using chromatin immunoprecipitation, we demonstrate that 2 of the CArG boxes, including the promoter-associated CArG box, bind to endogenous SRF in cultured aortic smooth muscle cells. Electrophoretic mobility-shift assays show that the conserved CArG boxes bind SRF in vitro. In reporter experiments, we show that the alternative promoter has transcriptional capacity that is dependent on SRF/myocardin and that the promoter associated CArG box is required for that activity. Finally, we show by quantitative RT-PCR that the alternative promoter is strongly downregulated in SRF-deficient embryonic stem cells and in smooth muscle tissues derived from conditional SRF knockout mice. Collectively, our data demonstrate that expression of LPP in smooth muscle is mediated by an alternative promoter that is regulated by SRF/myocardin.
Collapse
Affiliation(s)
- Marleen M.R. Petit
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Henrik Lindskog
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Erik Larsson
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Per Wasteson
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Elisabeth Athley
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Silke Breuer
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Meike Angstenberger
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - David Hertfelder
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Erney Mattsson
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Alfred Nordheim
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Sven Nelander
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Per Lindahl
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| |
Collapse
|
21
|
Lockman K, Taylor JM, Mack CP. The histone demethylase, Jmjd1a, interacts with the myocardin factors to regulate SMC differentiation marker gene expression. Circ Res 2007; 101:e115-23. [PMID: 17991879 DOI: 10.1161/circresaha.107.164178] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We and others have previously shown that the myocardin transcription factors play critical roles in the regulation of smooth muscle cell (SMC) differentiation marker gene expression. In a yeast 2-hybrid screen for proteins that interact with myocardin-related transcription factor-A (MRTF-A), we identified the histone 3 lysine 9 (H3K9)-specific demethylase, Jmjd1a. GST pull-down assays demonstrated that Jmjd1a bound all 3 myocardin family members, and further mapping studies showed that the jumonjiC domain of Jmjd1a was sufficient to mediate this interaction. Overexpression of Jmjd1a in multipotential 10T1/2 cells decreased global levels of di-methyl H3K9, stimulated the SM alpha-actin and SM22 promoters, and synergistically enhanced MRTF-A- and myocardin-dependent transactivation. Using chromatin immunoprecipitation assays, we also demonstrated that TGF-beta-mediated upregulation of SMC differentiation marker gene expression in 10T1/2 cells was associated with decreased H3K9 dimethylation at the CArG-containing regions of the SMC differentiation marker gene promoters. Importantly, knockdown of Jmjd1a in 10T1/2 cells and primary rat aortic SMCs by retroviral delivery of siRNA attenuated TGF-beta-induced upregulation of endogenous SM myosin heavy chain expression. These effects were concomitant with increased H3K9 dimethylation at the SMC differentiation marker gene promoters and with inhibition of MRTF-A-dependent transactivation of the SMC-specific transcription. These results suggest, for the first time, that SMC differentiation marker gene expression is regulated by H3K9 methylation and that the effects of the myocardin factors on SMC-specific transcription may involve the recruitment of Jmjd1a to the SMC-specific promoters.
Collapse
Affiliation(s)
- Kashelle Lockman
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599-7525, USA
| | | | | |
Collapse
|
22
|
Pidkovka NA, Cherepanova OA, Yoshida T, Alexander MR, Deaton RA, Thomas JA, Leitinger N, Owens GK. Oxidized Phospholipids Induce Phenotypic Switching of Vascular Smooth Muscle Cells In Vivo and In Vitro. Circ Res 2007; 101:792-801. [PMID: 17704209 DOI: 10.1161/circresaha.107.152736] [Citation(s) in RCA: 184] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atherosclerosis is a vascular disease characterized by lipid deposition and inflammation within the arterial wall. Oxidized phospholipids (oxPLs), such as 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (oxPAPC) and its constituents 1-palmytoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC) and 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphocholine (PGPC) are concentrated within atherosclerotic lesions and are known to be potent proinflammatory mediators. Phenotypic switching of smooth muscle cells (SMCs) plays a critical role in the development, progression, and end-stage clinical consequences of atherosclerosis, yet little is known regarding the effects of specific oxPLs on SMC phenotype. The present studies were focused on determining whether oxPLs regulate expression of SMC differentiation marker genes and the molecular mechanisms involved. Results showed that POVPC and PGPC induced profound suppression of smooth muscle (SM) α-actin and SM myosin heavy chain expression while simultaneously increasing expression of MCP-1, MCP-3, and cytolysin. OxPLs also induced nuclear translocation of Krüppel-like transcription factor 4 (KLF4), a known repressor of SMC marker genes. siRNA targeting of KLF4 nearly blocked POVPC-induced suppression of SMC marker genes, and myocardin. POVPC-induced repression of SMC marker genes was also significantly attenuated in KLF4 knockout SMCs. Taken together, these results suggest a novel role for oxPLs in phenotypic modulation of SMCs and indicate that these effects are dependent on the transcription factor, KLF4. These results may have important novel implications for the mechanisms by which oxPLs contribute to the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Nataliya A Pidkovka
- University of Virginia, Cardiovascular Research Center, Department of Molecular Physiology and Biophysics, 415 Lane Road, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Stepanek J, Vincent M, Turpin PY, Paulin D, Fermandjian S, Alpert B, Zentz C. C-->G base mutations in the CArG box of c-fos serum response element alter its bending flexibility. Consequences for core-SRF recognition. FEBS J 2007; 274:2333-48. [PMID: 17403043 DOI: 10.1111/j.1742-4658.2007.05768.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
By binding to the CArG box sequence, the serum response factor (SRF) activates several muscle-specific genes, as well as genes that respond to mitogens. The core domain of the SRF (core-SRF) binds as a dimer to the CArG box C-5C-4A-3T-2A-1T+1T+2A+3G+4G+5 of the c-fos serum response element (SREfos). However, previous studies using 20-mer DNAs have shown that the binding stoichiometry of core-SRF is significantly altered by mutations C-5-->G (SREGfos) and C-5C-4-->GG (SREGGfos) of the CArG box [A Huet, A Parlakian, M-C Arnaud, J-M Glandières, P Valat, S Fermandjian, D Paulin, B Alpert & C Zentz (2005) FEBS J272, 3105-3119]. To understand these effects, we carried out a comparative analysis of the three 20-mer DNAs SREfos, SREGfos and SREGGfos in aqueous solution. Their CD spectra were of the B-DNA type with small differences generated by variations in the mutual arrangement of the base pairs. Analysis by singular value decomposition of a set of Raman spectra recorded as a function of temperature, revealed a premelting transition associated with a conformational shift in the DNA double helices from a bent to a linear form. Time-resolved fluorescence anisotropy shows that the fluorescein reporter linked to the oligonucleotide 5'-ends experiences twisting motions of the double helices related to the interconversion between bent and linear conformers. The three SREs present various bent populations submitted, however, to particular internal dynamics, decisive for the mutual adjustment of binding partners and therefore specific complex formation.
Collapse
Affiliation(s)
- Josef Stepanek
- Laboratoire de Biophysique Moléculaire Cellulaire & Tissulaire, Université Pierre et Marie Curie, Evry, France
| | | | | | | | | | | | | |
Collapse
|
24
|
Hinson JS, Medlin MD, Lockman K, Taylor JM, Mack CP. Smooth muscle cell-specific transcription is regulated by nuclear localization of the myocardin-related transcription factors. Am J Physiol Heart Circ Physiol 2007; 292:H1170-80. [PMID: 16997888 DOI: 10.1152/ajpheart.00864.2006] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
On the basis of our previous studies on RhoA signaling in smooth muscle cells (SMC), we hypothesized that RhoA-mediated nuclear translocalization of the myocardin-related transcription factors (MRTFs) was important for regulating SMC phenotype. MRTF-A protein and MRTF-B message were detected in aortic SMC and in many adult mouse organs that contain a large SMC component. Both MRTFs upregulated SMC-specific promoter activity as well as endogenous SM22α expression in multipotential 10T1/2 cells, although to a lesser extent than myocardin. We used enhanced green fluorescent protein (EGFP) fusion proteins to demonstrate that the myocardin factors have dramatically different localization patterns and that the stimulation of SMC-specific transcription by certain RhoA-dependent agonists was likely mediated by increased nuclear translocation of the MRTFs. Importantly, a dominant-negative form of MRTF-A (ΔB1/B2) that traps endogenous MRTFs in the cytoplasm inhibited the SM α-actin, SM22α, and SM myosin heavy chain promoters in SMC and attenuated the effects of sphingosine 1-phosphate and transforming growth factor (TGF)-β on SMC-specific transcription. Our data confirmed the importance of the NH2-terminal RPEL domains for regulating MRTF localization, but our analysis of MRTF-A/myocardin chimeras and myocardin RPEL2 mutations indicated that the myocardin B1/B2 region can override this signal. Gel shift assays demonstrated that myocardin factor activity correlated well with ternary complex formation at the SM α-actin CArGs and that MRTF-serum response factor interactions were partially dependent on CArG sequence. Taken together, our results indicate that the MRTFs regulate SMC-specific gene expression in at least some SMC subtypes and that regulation of MRTF nuclear localization may be important for the effects of selected agonists on SMC phenotype.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Animals
- Aorta, Thoracic/metabolism
- Cell Differentiation
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Lysophospholipids/pharmacology
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Mutation
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Platelet-Derived Growth Factor/pharmacology
- Promoter Regions, Genetic/drug effects
- RNA, Messenger/metabolism
- Rats
- Serum Response Factor/metabolism
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription, Genetic/drug effects
- Transfection
- Transforming Growth Factor beta/pharmacology
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Jeremiah S Hinson
- Department of Pathology and Laboratory Medicine and the Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | |
Collapse
|
25
|
Staus DP, Blaker AL, Taylor JM, Mack CP. Diaphanous 1 and 2 regulate smooth muscle cell differentiation by activating the myocardin-related transcription factors. Arterioscler Thromb Vasc Biol 2006; 27:478-86. [PMID: 17170370 DOI: 10.1161/01.atv.0000255559.77687.c1] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We have previously shown that smooth muscle cell (SMC) differentiation marker gene expression is regulated by the small GTPase, RhoA. The objective of the present study was to determine the contributions of the RhoA effectors, diaphanous 1 and 2 (mDia1 and mDia2), to this regulatory mechanism. METHODS AND RESULTS mDia1 and mDia2 are expressed highly in aortic SMCs and in a number of SMC-containing organs including bladder, lung, and esophagus. Activation of mDia1/2 signaling by RhoA strongly stimulated SMC-specific promoter activity in multiple cell-types including primary aortic SMCs, and stimulated endogenous SM alpha-actin expression in 10T1/2 cells. Expression of a dominant negative Dia1 variant that inhibits both mDia1 and mDia2 significantly decreased SMC-specific transcription in SMCs. The effects of mDia1 and mDia2 required the presence of SRF and the activity of the myocardin transcription factors and were dependent on changes in actin polymerization. Importantly, stimulation of mDia1/2 signaling synergistically enhanced the activities of the myocardin-related transcription factors, MRTF-A and MRTF-B, and this effect was attributable to increased nuclear localization of these factors. CONCLUSIONS These results indicate that RhoA-dependent signaling through mDia1/2 and the MRTFs is important for SMC-specific gene expression in SMCs.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Blotting, Western
- Carrier Proteins/metabolism
- Carrier Proteins/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cells, Cultured
- Formins
- Gene Expression Regulation
- Immunohistochemistry
- Mice
- Models, Animal
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Nuclear Proteins/metabolism
- Rats
- Reverse Transcriptase Polymerase Chain Reaction
- Sensitivity and Specificity
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Signal Transduction/physiology
- Trans-Activators/metabolism
- Transcription Factors/metabolism
- Transcription, Genetic
- rhoA GTP-Binding Protein/genetics
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Dean P Staus
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525, USA
| | | | | | | |
Collapse
|
26
|
Mack CP, Hinson JS. Regulation of smooth muscle differentiation by the myocardin family of serum response factor co-factors. J Thromb Haemost 2005; 3:1976-84. [PMID: 15892867 DOI: 10.1111/j.1538-7836.2005.01316.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- C P Mack
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | |
Collapse
|
27
|
Matsumura T, Suzuki T, Aizawa K, Munemasa Y, Muto S, Horikoshi M, Nagai R. The deacetylase HDAC1 negatively regulates the cardiovascular transcription factor Krüppel-like factor 5 through direct interaction. J Biol Chem 2005; 280:12123-9. [PMID: 15668237 DOI: 10.1074/jbc.m410578200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transcription is regulated by a network of transcription factors and related cofactors that act in concert with the general transcription machinery. Elucidating their underlying interactions is important for understanding the mechanisms regulating transcription. Recently, we have shown that Krüppel-like factor KLF5, a member of the Sp/KLF family of zinc finger factors and a key regulator of cardiovascular remodeling, is regulated positively by the acetylase p300 and negatively by the oncogenic regulator SET through coupled interaction and regulation of acetylation. Here, we have shown that the deacetylase HDAC1 can negatively regulate KLF5 through direct interaction. KLF5 interacts with HDAC1 in the cell and in vitro. Gel shift DNA binding assay showed that their interaction inhibits the DNA binding activity of KLF5, suggesting a property of HDAC1 to directly affect the DNA binding affinity of a transcription factor. Reporter assay also revealed that HDAC1 suppresses KLF5-dependent promoter activation. Additionally, overexpression of HDAC1 suppressed KLF5-dependent activation of its endogenous downstream gene, platelet-derived growth factor-A chain gene, when activated by phorbol ester. Further, HDAC1 binds to the first zinc finger of KLF5, which is the same region where p300 interacts with KLF5 and, intriguingly, HDAC1 inhibits binding of p300 to KLF5. Direct competitive interaction between acetylase and deacetylase has been hitherto unknown. Collectively, the transcription factor KLF5 is negatively regulated by the deacetylase HDAC1 through direct effects on its activities (DNA binding activity, promoter activation) and further through inhibition of interaction with p300. These findings suggest a novel role and mechanism for regulation of transcription by deacetylase.
Collapse
Affiliation(s)
- Takayoshi Matsumura
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Stolle K, Weitkamp B, Rauterberg J, Lorkowski S, Cullen P. Laser microdissection-based analysis of mRNA expression in human coronary arteries with intimal thickening. J Histochem Cytochem 2004; 52:1511-8. [PMID: 15505346 PMCID: PMC3957817 DOI: 10.1369/jhc.4a6289.2004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2004] [Accepted: 05/17/2004] [Indexed: 11/22/2022] Open
Abstract
Intimal thickening is an early phase of atherosclerosis characterized by differentiation of plaque smooth muscle cells (SMCs) from a contractile to a synthetic phenotype. We used laser microdissection (LMD) plus real-time RT-PCR to quantify mRNAs for calponin-1 and smoothelin, markers of the contractile phenotype, and for serum response factor (SRF), a regulator of SMC differentiation, in intimal and medial SMCs of human coronary arteries with intimal thickening. RNA expression was also analyzed by ISH and protein expression was detected by IHC. LMD plus RT-PCR found similar levels of SRF mRNA in intimal and medial SMCs, while medial mRNA levels for calponin-1 and smoothelin were higher. ISH confirmed that smoothelin mRNA levels in media exceeded those in intima, whereas SRF mRNA levels were similar at both sites. For calponin-1 and smoothelin, protein levels mirrored respective mRNA levels. By contrast, more medial than intimal SRF protein was present. Our results indicate that intimal SMCs exhibit a largely synthetic phenotype, perhaps reflecting lower intimal levels of SRF protein; ISH and LMD plus real-time RT-PCR provide comparable results; as a valuable alternative to ISH, LMD plus RT-PCR allows parallel measurement of several transcripts; and tissue gene expression studies must measure both protein and mRNA levels.
Collapse
Affiliation(s)
- Katrin Stolle
- Institute of Arteriosclerosis Research (KS, BW, JR, SL, PC), University of Münster, Münster, Germany
- Institute of Biochemistry (KS, SL), University of Münster, Münster, Germany
| | - Benedikt Weitkamp
- Institute of Arteriosclerosis Research (KS, BW, JR, SL, PC), University of Münster, Münster, Germany
| | - Jürgen Rauterberg
- Institute of Arteriosclerosis Research (KS, BW, JR, SL, PC), University of Münster, Münster, Germany
| | - Stefan Lorkowski
- Institute of Arteriosclerosis Research (KS, BW, JR, SL, PC), University of Münster, Münster, Germany
- Institute of Biochemistry (KS, SL), University of Münster, Münster, Germany
| | - Paul Cullen
- Institute of Arteriosclerosis Research (KS, BW, JR, SL, PC), University of Münster, Münster, Germany
| |
Collapse
|
29
|
Wamhoff BR, Hoofnagle MH, Burns A, Sinha S, McDonald OG, Owens GK. A G/C element mediates repression of the SM22alpha promoter within phenotypically modulated smooth muscle cells in experimental atherosclerosis. Circ Res 2004; 95:981-8. [PMID: 15486317 DOI: 10.1161/01.res.0000147961.09840.fb] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A hallmark of smooth muscle cell (SMC) phenotypic switching in atherosclerotic lesions is suppression of SMC differentiation marker gene expression. Yet little is known regarding the molecular mechanisms that control this process. Here we show that transcription of the SMC differentiation marker gene SM22alpha is reduced in atherosclerotic lesions and identify a cis regulatory element in the SM22alpha promoter required for this process. Transgenic mice carrying the SM22alpha promoter-beta-galactosidase (beta-gal) reporter transgene were crossed to apolipoprotein E (ApoE)-/- mice. Cells of the fibrous cap, intima, and underlying media showed complete loss of beta-gal activity in advanced atherosclerotic lesions. Of major significance, mutation of a G/C-rich cis element in the SM22alpha promoter prevented the decrease in SM22alpha promoter-beta-gal reporter transgene expression, including in cells that compose the fibrous cap of the lesion and in medial cells in proximity to the lesion. To begin to assess mechanisms whereby the G/C repressor element mediates suppression of SM22alpha in atherosclerosis, we tested the hypothesis that effects may be mediated by platelet-derived growth factor (PDGF)-BB-induced increases in the G/C binding transcription factor Sp1. Consistent with this hypothesis, results of studies in cultured SMCs showed that: (1) PDGF-BB increased expression of Sp1; (2) PDGF-BB and Sp1 profoundly suppressed SM22alpha promoter activity as well as smooth muscle myosin heavy chain promoter activity through mechanisms that were at least partially dependent on the G/C cis element; and (3) a short interfering RNA to Sp1 increased basal expression and attenuated PDGF-BB induced suppression of SM22alpha. Together, these results support a model whereby a G/C repressor element within the SM22alpha promoter mediates transcriptional repression of this gene within phenotypically modulated SMCs in experimental atherosclerosis and provide indirect evidence implicating PDGF-BB and Sp1 as possible mediators of these effects.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Apolipoproteins E/genetics
- Arteriosclerosis/etiology
- Arteriosclerosis/genetics
- Arteriosclerosis/pathology
- Becaplermin
- Cell Differentiation/genetics
- Cells, Cultured/metabolism
- Cells, Cultured/pathology
- Crosses, Genetic
- Extracellular Matrix Proteins/biosynthesis
- Extracellular Matrix Proteins/genetics
- Gene Silencing/physiology
- Genes, Reporter
- Hypercholesterolemia/complications
- Hypercholesterolemia/genetics
- Lac Operon
- Mice
- Mice, Inbred CBA
- Mice, Transgenic
- Microfilament Proteins/genetics
- Muscle Proteins/biosynthesis
- Muscle Proteins/genetics
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Platelet-Derived Growth Factor/pharmacology
- Platelet-Derived Growth Factor/physiology
- Promoter Regions, Genetic/genetics
- Protein Binding
- Proto-Oncogene Proteins c-sis
- Rats
- Recombinant Fusion Proteins/physiology
- Regulatory Sequences, Nucleic Acid
- Serum Response Element
- Sp1 Transcription Factor/physiology
Collapse
Affiliation(s)
- B R Wamhoff
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville 22908-0736, USA
| | | | | | | | | | | |
Collapse
|
30
|
Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev 2004; 84:767-801. [PMID: 15269336 DOI: 10.1152/physrev.00041.2003] [Citation(s) in RCA: 2624] [Impact Index Per Article: 125.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The focus of this review is to provide an overview of the current state of knowledge of molecular mechanisms/processes that control differentiation of vascular smooth muscle cells (SMC) during normal development and maturation of the vasculature, as well as how these mechanisms/processes are altered in vascular injury or disease. A major challenge in understanding differentiation of the vascular SMC is that this cell can exhibit a wide range of different phenotypes at different stages of development, and even in adult organisms the cell is not terminally differentiated. Indeed, the SMC is capable of major changes in its phenotype in response to changes in local environmental cues including growth factors/inhibitors, mechanical influences, cell-cell and cell-matrix interactions, and various inflammatory mediators. There has been much progress in recent years to identify mechanisms that control expression of the repertoire of genes that are specific or selective for the vascular SMC and required for its differentiated function. One of the most exciting recent discoveries was the identification of the serum response factor (SRF) coactivator gene myocardin that appears to be required for expression of many SMC differentiation marker genes, and for initial differentiation of SMC during development. However, it is critical to recognize that overall control of SMC differentiation/maturation, and regulation of its responses to changing environmental cues, is extremely complex and involves the cooperative interaction of many factors and signaling pathways that are just beginning to be understood. There is also relatively recent evidence that circulating stem cell populations can give rise to smooth muscle-like cells in association with vascular injury and atherosclerotic lesion development, although the exact role and properties of these cells remain to be clearly elucidated. The goal of this review is to summarize the current state of our knowledge in this area and to attempt to identify some of the key unresolved challenges and questions that require further study.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Arteriosclerosis/genetics
- Cell Differentiation
- Cellular Senescence
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Vascular Diseases/genetics
- Vascular Diseases/metabolism
- Vascular Diseases/pathology
Collapse
Affiliation(s)
- Gary K Owens
- Dept. of Molecular Physiology and Biological Physics, Univ. of Virginia School of Medicine, 415 Lane Rd., Medical Research Building 5, Rm. 1220, PO Box 801394, Charlottesville, VA 22908, USA.
| | | | | |
Collapse
|
31
|
Novak EM, Metzger M, Chammas R, da Costa M, Dantas K, Manabe C, Pires J, de Oliveira AC, Bydlowski SP. Downregulation of TNF-alpha and VEGF expression by Sp1 decoy oligodeoxynucleotides in mouse melanoma tumor. Gene Ther 2003; 10:1992-7. [PMID: 14528323 DOI: 10.1038/sj.gt.3302111] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Melanoma tumor growth and progression are highly dependent on adequate blood supply through angiogenesis. Since several genes involved in angiogenesis revealed potential binding sites for the transcription factor Sp1, we have examined the effects of local inoculation of Sp1 decoy oligodeoxynucleotides (ODNs) on the growth of transplanted murine melanoma tumors and the expression of VEGF and TNF-alpha within these tumors. Treatment with Sp1 decoy ODNs, but not their mutated form, led to a significant increase (P=0.041) of the tumor necrotic area, as evaluated morphometrically. Tumor necrosis was associated with a significant decrease of microvascular density (P=0.012) and relative vascular area (P=0.026), as determined by counting CD34-positive vascular structures within the tumor microenvironment of Sp1 decoy ODNs and control ODN-treated tumors. RT-PCR experiments showed a strong decrease in the levels of VEGF188 and VEGF164 isoforms and a moderate decrease of TNF-alpha in Sp1 decoy-treated tumors. Taken together, our results indicate that Sp1 decoy ODNs may inhibit angiogenesis by affecting the gene expression of key players in angiogenesis such as TNF-alpha and VEGF. These findings indicate that Sp1 decoy ODNs may be a potential new therapeutic tool in antiangiogenic therapy.
Collapse
Affiliation(s)
- E M Novak
- Research and Molecular Biology Division, Pró-Sangue Hemocentro de São Paulo Foundation, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chang YF, Wei J, Liu X, Chen YH, Layne MD, Yet SF. Identification of a CArG-independent region of the cysteine-rich protein 2 promoter that directs expression in the developing vasculature. Am J Physiol Heart Circ Physiol 2003; 285:H1675-83. [PMID: 12791591 DOI: 10.1152/ajpheart.00165.2003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cysteine-rich protein (CRP)2 is a member of the LIM-only CRP family that is expressed in vascular smooth muscle cells (VSMC). To gain insight into the transcription of CSRP2 (gene name for CRP2) in VSMC, we analyzed the 5'-flanking sequence of the CSRP2 gene. We showed previously that 4,855 bp of the 5'-flanking sequence of the CSRP2 gene directed lacZ reporter gene expression, primarily in the VSMC of transgenic mice. To further define the regulatory sequences important for CSRP2 expression in VSMC, a series of promoter constructs containing deletions of the 5'-flanking sequence upstream of a nuclear-localized lacZ reporter gene were generated and analyzed. Similar to that observed in the -4855CSRP2-lacZ mice, beta-galactosidase reporter activity was detected in the developing great vessels, aorta, intersegmental arteries, umbilical vessels, endocardial cushions, and neural tube in the -3513-, -2663-, -795-, and -664CSRP2-lacZ lines. However, an internal deletion of bp -573 to -550 abolished the vascular, but not the neural tube, staining. Interestingly, no CArG box [CC(A/T)6GG] was present in the -795-bp fragment. Cotransfection experiments showed that dominant-negative serum response factor (SRF) did not repress CSRP2 promoter activity, which was different from the repressive effect of dominant-negative SRF on the SM22 alpha promoter. Our data suggest the presence of a VSMC-specific element(s) within bp -573 to -550 of the CSRP2 5'-flanking sequence; however, in contrast to many other smooth muscle genes, transcriptional regulation of the CSRP2 gene is not dependent on SRF.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Base Sequence/genetics
- Blood Vessels/embryology
- Blood Vessels/growth & development
- Blood Vessels/metabolism
- Cell Cycle Proteins
- Cells, Cultured
- DNA-Binding Proteins/physiology
- Embryo, Mammalian/metabolism
- Embryonic and Fetal Development
- Gene Expression
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B
- Male
- Mice
- Mice, Transgenic
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/physiology
- Protein Structure, Tertiary/genetics
- Proteins
- Rats
- Rats, Sprague-Dawley
- Repressor Proteins/physiology
- Ribonucleoproteins
- Transcription Factors
- Transgenes
Collapse
Affiliation(s)
- Yung-Fu Chang
- Pulmonary and Critical Care Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
33
|
Appleby CE, Kingston PA, David A, Gerdes CA, Umaña P, Castro MG, Lowenstein PR, Heagerty AM. A novel combination of promoter and enhancers increases transgene expression in vascular smooth muscle cells in vitro and coronary arteries in vivo after adenovirus-mediated gene transfer. Gene Ther 2003; 10:1616-22. [PMID: 12907954 PMCID: PMC2902242 DOI: 10.1038/sj.gt.3302044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2003] [Accepted: 02/25/2003] [Indexed: 11/09/2022]
Abstract
Recombinant adenoviruses are employed widely for vascular gene transfer. Vascular smooth muscle cells (SMCs) are a relatively poor target for transgene expression after adenovirus-mediated gene delivery, however, even when expression is regulated by powerful, constitutive viral promoters. The major immediate-early murine cytomegalovirus enhancer/promoter (MIEmCMV) elicits substantially greater transgene expression than the human cytomegalovirus promoter (MIEhCMV) in all cell types in which they have been compared. The Woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) increases transgene expression in numerous cell lines, and fragments of the smooth muscle myosin heavy chain (SMMHC) promoter increase expression within SMC from heterologous promoters. We therefore, compared the expression of beta-galactosidase after adenovirus-mediated gene transfer of lacZ under the transcriptional regulation of a variety of combinations of the promoters and enhancers described, in vitro and in porcine coronary arteries. We demonstrate that inclusion of WPRE and a fragment of the rabbit SMMHC promoter along with MIEmCMV increases beta-galactosidase expression 90-fold in SMC in vitro and approximately 40-fold in coronary arteries, compared with vectors in which expression is regulated by MIEhCMV alone. Expression cassette modification represents a simple method of improving adenovirus-mediated vascular gene transfer efficiency and has important implications for the development of efficient cardiovascular gene therapy strategies.
Collapse
|
34
|
Zhang J, Wang S, Wesley RA, Danner RL. Adjacent sequence controls the response polarity of nitric oxide-sensitive Sp factor binding sites. J Biol Chem 2003; 278:29192-200. [PMID: 12759366 DOI: 10.1074/jbc.m213043200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Nitric oxide (NO*) and cAMP-dependent protein kinase (PKA) inhibitors up-regulate tumor necrosis factor alpha (TNFalpha) by decreasing Sp1 binding to a proximal GC box element. Here, elements flanking GC boxes were tested for their role in determining whether Sp sites act as activators or repressors. Promoter studies in receptive human cell lines demonstrated that NO* down-regulated endothelial NO* synthase (eNOS) but up-regulated TNFalpha. Like TNFalpha, Sp1 binding to the eNOS promoter was decreased by NO* and a PKA inhibitor, H89, and increased by a PKA activator, dibutyryl cAMP (Bt2cAMP). For either promoter, mutation of Sp sites abolished NO* responses. In contrast, mutation of an upstream AP1 site in the TNFalpha promoter (not present in eNOS) maintained NO* responsiveness, but reversed the direction of NO* and cAMP effects. Using artificial constructs, NO* increased transcription when Sp and AP1 sites were both present (TNFalpha-like response), but decreased it when the adjacent AP1 site was disrupted (eNOS-like response). NO*, H89, and Bt2cAMP were found to produce reciprocal protein binding changes at contiguous AP1 and Sp sites (p < 0.0001 for an interaction). Chromatin immunoprecipitation assays demonstrated that Sp1 and to a lesser extent Sp3 bound to the GC box regions of eNOS and TNFalpha in intact cells. Thus, this NO*- and cAMP-responsive regulatory module has a Sp site sensor variably coupled to an adjacent element that determines response polarity. These results define a composite element that can utilize secondary inputs to convert off signals to on, thereby conferring complex functionalities to the same DNA binding motif.
Collapse
Affiliation(s)
- Jianhua Zhang
- Critical Care Medicine Department, Warren Grant Magnuson Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
35
|
Liu HW, Halayko AJ, Fernandes DJ, Harmon GS, McCauley JA, Kocieniewski P, McConville J, Fu Y, Forsythe SM, Kogut P, Bellam S, Dowell M, Churchill J, Lesso H, Kassiri K, Mitchell RW, Hershenson MB, Camoretti-Mercado B, Solway J. The RhoA/Rho kinase pathway regulates nuclear localization of serum response factor. Am J Respir Cell Mol Biol 2003; 29:39-47. [PMID: 12600823 DOI: 10.1165/rcmb.2002-0206oc] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
RhoA and its downstream target Rho kinase regulate serum response factor (SRF)-dependent skeletal and smooth muscle gene expression. We previously reported that long-term serum deprivation reduces transcription of smooth muscle contractile apparatus encoding genes, by redistributing SRF out of the nucleus. Because serum components stimulate RhoA activity, these observations suggest the hypothesis that the RhoA/Rho kinase pathway regulates SRF-dependent smooth muscle gene transcription in part by controlling SRF subcellular localization. Our present results support this hypothesis: cotransfection of cultured airway myocytes with a plasmid expressing constitutively active RhoAV14 selectively enhanced transcription from the SM22 and smooth muscle myosin heavy chain promoters and from a purely SRF-dependent promoter, but had no effect on transcription from the MSV-LTR promoter or from an AP2-dependent promoter. Conversely, inhibition of the RhoA/Rho kinase pathway by cotransfection with a plasmid expressing dominant negative RhoAN19, by cotransfection with a plasmid expressing Clostridial C3 toxin, or by incubation with the Rho kinase inhibitor, Y-27632, all selectively reduced SRF-dependent smooth muscle promoter activity. Furthermore, treatment with Y-27632 selectively reduced binding of SRF from nuclear extracts to its consensus DNA target, selectively reduced nuclear SRF protein content, and partially redistributed SRF from nucleus to cytoplasm, as revealed by quantitative immunocytochemistry. Treatment of cultured airway myocytes with latrunculin B, which reduces actin polymerization, also caused partial redistribution of SRF into the cytoplasm. Together, these results demonstrate for the first time that the RhoA/Rho kinase pathway controls smooth muscle gene transcription in differentiated smooth muscle cells, in part by regulating the subcellular localization of SRF. It is conceivable that the RhoA/Rho kinase pathway influences SRF localization through its effect on actin polymerization dynamics.
Collapse
Affiliation(s)
- Hong Wei Liu
- Departments of Medicine and Pediatrics, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Schauwienold D, Plum C, Helbing T, Voigt P, Bobbert T, Hoffmann D, Paul M, Reusch HP. ERK1/2-dependent contractile protein expression in vascular smooth muscle cells. Hypertension 2003; 41:546-52. [PMID: 12623957 DOI: 10.1161/01.hyp.0000054213.37471.84] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In vivo, vascular smooth muscle (VSM) cells change their contractile phenotype toward a more proliferative phenotype during the pathogenesis of vascular diseases. Because these dedifferentiated VSM cells may gradually regain contractile functions, we aimed to identify signaling pathways that result in an increased expression of contractile proteins in VSM cells. In vitro, serum and thrombin induced a reversible upregulation of smooth muscle myosin heavy-chain (SM-MHC) in cultured neonatal rat VSM cells. Cotransfection of a SM-MHC-promoter chloramphenicol acetyltransferase-construct with dominant-negative N17Ras or N17Raf or treatment with the mitogen-activated/ERK-activating kinase (MEK) inhibitor PD 98059 concentration dependently decreased the serum- or thrombin-induced SM-MHC promoter activity. Consistently, the serum- or thrombin-induced phosphorylation of MEK and extracellular signal-regulated kinase 1/2 (ERK1/2) coincided with a MEK-dependent nuclear accumulation of phosphorylated ERK1/2 and subsequent nuclear phosphorylation of the transcription factors c-myc and Elk-1. A 5'-deletion analysis of cis-elements within the SM-MHC promoter demonstrated that a conserved region (nucleotide -1346 to -1102) was required for both cell type-specific expression and serum- or thrombin-induced upregulation of the SM-MHC promoter in VSM cells. Within this region, 2 CArG-boxes, a GC-rich element, and a CTF/NF-1 site are critical positively acting cis-elements for the serum- or thrombin-induced upregulation of SM-MHC. We conclude that the serum- or thrombin-induced differentiation requires an intact Ras/Raf/MEK/ERK signaling cascade, nuclear translocation of activated ERK1/2, phosphorylation of transcription factors, and several cis-elements within the SM-MHC promoter.
Collapse
Affiliation(s)
- Dag Schauwienold
- Institut für Klinische Pharmakologie und Toxikologie, Freie Universität Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abe M, Hasegawa K, Wada H, Morimoto T, Yanazume T, Kawamura T, Hirai M, Furukawa Y, Kita T. GATA-6 is involved in PPARgamma-mediated activation of differentiated phenotype in human vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2003; 23:404-10. [PMID: 12615657 DOI: 10.1161/01.atv.0000059405.51042.a0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a member of the nuclear receptor superfamily involved in the growth and differentiation of many cell types. Although the activation of PPARgamma in human vascular smooth muscle cells (VSMCs) inhibits the growth of these cells, the precise mechanism of this effect is unknown. PPARgamma-mediated growth inhibition of VSMCs is associated with the induction of the differentiated phenotype. A zinc finger transcription factor, GATA-6, has been implicated in the maintenance of the differentiated phenotype in VSMCs. METHODS AND RESULTS The administration of 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), a naturally occurring PPARgamma ligand, and troglitazone, a thiazolidinedione derivative, induced the expression of smooth muscle myosin heavy chain and smooth muscle alpha-actin, highly specific markers for differentiated VSMCs. Stimulation of proliferative VSMCs with PPARgamma ligands also increased the activity of the transfected wild-type smooth muscle myosin heavy chain promoter but not that of the mutant promoter, in which a GATA-6 binding site was mutated. Compatible with the role of GATA-6, both 15d-PGJ2 and troglitazone upregulated the DNA binding activity of GATA-6 in proliferative VSMCs. CONCLUSIONS The activation of PPARgamma-dependent pathways induces the differentiated phenotype in proliferative VSMCs, and this induction is mediated, in part, through a GATA-6-dependent transcriptional mechanism.
Collapse
Affiliation(s)
- Mitsuru Abe
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Miano JM. Mammalian smooth muscle differentiation: origins, markers and transcriptional control. Results Probl Cell Differ 2003; 38:39-59. [PMID: 12132398 DOI: 10.1007/978-3-540-45686-5_2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Joseph M Miano
- Center for Cardiovascular Research, Box 679, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York 14642, USA
| |
Collapse
|
39
|
Layne MD, Yet SF, Maemura K, Hsieh CM, Liu X, Ith B, Lee ME, Perrella MA. Characterization of the mouse aortic carboxypeptidase-like protein promoter reveals activity in differentiated and dedifferentiated vascular smooth muscle cells. Circ Res 2002; 90:728-36. [PMID: 11934842 DOI: 10.1161/01.res.0000013289.97650.c8] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The dedifferentiation and proliferation of vascular smooth muscle cells (VSMCs) contribute to the formation of vascular lesions. In this study, the regulation of aortic carboxypeptidase-like protein (ACLP) expression in VSMCs was investigated. After mouse carotid injury, the expression of ACLP increases in the dedifferentiated VSMCs of the neointima in a pattern that differs from that of smooth muscle alpha-actin. To better understand the regulation of ACLP in VSMCs, we characterized the 21-exon mouse ACLP gene and 5'-flanking region and examined its promoter activity. In transient transfection assays, 2.5 kb of the ACLP 5'-flanking sequence directed high levels of luciferase reporter activity in primary cultured rat aortic smooth muscle cells, and this activity was not dependent on serum response factor. We identified a positive element between base pairs -156 and -122 by analysis of 5' deletion and mutant constructs. By use of electrophoretic mobility shift assays with rat aortic smooth muscle cell nuclear extracts, Sp1 and Sp3 transcription factors bound to this region, and transfection assays in D.Mel.2 cells revealed that both Sp1 and Sp3 transactivated the ACLP promoter. Transgenic mice harboring the -2.5-kb ACLP promoter upstream from a nuclear-targeted LacZ gene were generated, and expression was detected in the VSMCs of large blood vessels, arterioles, and veins. Interestingly, ACLP promoter-LacZ reporter activity increased within the neointimal VSMCs of injured carotid vessels, consistent with the expression of the endogenous ACLP protein. The ACLP promoter may provide a novel tool to target gene expression to dedifferentiated VSMCs.
Collapse
Affiliation(s)
- Matthew D Layne
- Pulmonary and Critical Care, Divisions and the Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Wada H, Hasegawa K, Morimoto T, Kakita T, Yanazume T, Abe M, Sasayama S. Calcineurin-GATA-6 pathway is involved in smooth muscle-specific transcription. J Cell Biol 2002; 156:983-91. [PMID: 11889139 PMCID: PMC2173472 DOI: 10.1083/jcb.200106057] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intracellular calcium is one of the important signals that initiates the myogenic program. The calcium-activated phosphatase calcineurin is necessary for the nuclear import of the nuclear factor of activated T cell (NFAT) family members, which interact with zinc finger GATA transcription factors. Whereas GATA-6 plays a role in the maintenance of the differentiated phenotype in vascular smooth muscle cells (VSMCs), it is unknown whether the calcineurin pathway is associated with GATA-6 and plays a role in the differentiation of VSMCs. The smooth muscle-myosin heavy chain (Sm-MHC) gene is a downstream target of GATA-6, and provides a highly specific marker for differentiated VSMCs. Using immunoprecipitation Western blotting, we showed that NFATc1 interacted with GATA-6. Consistent with this, NFATc1 further potentiated GATA-6-activated Sm-MHC transcription. Induction of VSMCs to the quiescent phenotype caused nuclear translocation of NFATc1. In differentiated VSMCs, blockage of calcineurin down-regulated the amount of GATA-6-DNA binding as well as the expression of Sm-MHC and its transcriptional activity. These findings demonstrate that the calcineurin pathway is associated with GATA-6 and is required for the maintenance of the differentiated phenotype in VSMCs.
Collapse
MESH Headings
- Actins/drug effects
- Actins/genetics
- Actins/metabolism
- Animals
- COS Cells
- Calcineurin/drug effects
- Calcineurin/genetics
- Calcineurin/metabolism
- Calcium Signaling/drug effects
- Calcium Signaling/physiology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cell Division/drug effects
- Cell Division/physiology
- Cell Nucleus/drug effects
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Common Variable Immunodeficiency
- DNA/drug effects
- DNA/metabolism
- DNA-Binding Proteins/drug effects
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- GATA6 Transcription Factor
- Humans
- Immunohistochemistry
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/metabolism
- Myosin Heavy Chains/drug effects
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- NFATC Transcription Factors
- Nuclear Proteins
- Phenotype
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/physiology
- Protein Transport/drug effects
- Protein Transport/genetics
- Transcription Factors/drug effects
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic/drug effects
- Transcription, Genetic/physiology
Collapse
Affiliation(s)
- Hiromichi Wada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Nagai R, Suzuki T, Aizawa K, Miyamoto S, Amaki T, Kawai-Kowase K, Sekiguchi KI, Kurabayashi M. Phenotypic modulation of vascular smooth muscle cells: dissection of transcriptional regulatory mechanisms. Ann N Y Acad Sci 2001; 947:56-66; discussion 66-7. [PMID: 11795310 DOI: 10.1111/j.1749-6632.2001.tb03930.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The smooth muscle myosin heavy chain (MHC) gene and its isoforms are excellent molecular markers that reflect smooth muscle phenotypes. The SMemb/Nonmuscle Myosin Heavy Chain B (NMHC-B) is a distinct MHC gene expressed predominantly in phenotypically modulated SMCs (synthetic-type SMC). To dissect the molecular mechanisms governing phenotypic modulation of SMCs, we analyzed the transcriptional regulatory mechanisms underlying expression of the SMemb gene. We previously reported two transcription factors, BTEB2/IKLF and Hex, which transactivate the SMemb gene promoter based on the transient reporter transfection assays. BTEB2/IKLF is a zinc finger transcription factor, whereas Hex is a homeobox protein. BTEB2/IKLF expression in SMCs is downregulated with vascular development in vivo but upregulated in cultured SMCs and in neointima in response to vascular injury after balloon angioplasty. BTEB2/IKLF and Hex activate not only the SMemb gene but also other genes activated in synthetic SMCs including plasminogen activator inhibitor-1 (PAI-1), iNOS, PDGF-A, Egr-1, and VEGF receptors. Mitogenic stimulation activates BTEB2/IKLF gene expression through MEK1 and Egr-1. Elevation of intracellular cAMP is also important in phenotypic modulation of SMCs, because the SMemb promoter is activated under cooperatively by cAMP-response element binding protein (CREB) and Hex.
Collapse
Affiliation(s)
- R Nagai
- Department of Cardiovascular Medicine, University of Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Manabe I, Owens GK. The smooth muscle myosin heavy chain gene exhibits smooth muscle subtype-selective modular regulation in vivo. J Biol Chem 2001; 276:39076-87. [PMID: 11489897 DOI: 10.1074/jbc.m105402200] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous studies in our laboratory demonstrated that the transgene consisting of the -4.2 to +11.6 kilobase (kb) region of the smooth muscle (SM) myosin heavy chain (MHC) gene was expressed in virtually all SM tissue types in vivo in transgenic mice and that the multiple CArG elements within this region were differentially required in SMC subtypes, implying that the SM-MHC gene was controlled by multiple transcriptional regulatory modules. To investigate this hypothesis, we analyzed specific regulatory regions within the SM-MHC -4.2 to +11.6 kb region by a combination of deletion analyses of various SM-MHC transgenes as well as by DNaseI hypersensitivity assays and in vivo footprinting in intact SMC tissues. The results showed that SM-MHC transgene expression depended on a large number of required regulatory modules that were widely spread over the -4.2 to +11.6 region. Moreover, the results revealed several unexpected novel features of regulation of the SM-MHC gene including: 1) unique combinations of regulatory modules were required for SM-MHC expression in different SMC-subtypes; 2) repressor modules as well as activator modules were both critical for SMC specificity of the gene; 3) certain modules were required in certain contexts but were dispensable in others within a given SMC-subtype (i.e. the net activity of the module was determined by interaction between modules not simply by the sum of module activities); and 4) we identified a highly conserved 200-base pair transcriptional regulatory module at +8 kb that was required in the large arteries but dispensable in the coronary arteries and airways in transgenic mice and contained multiple potential cis-elements that were occupied by nuclear proteins in the intact aorta based on in vivo footprinting. Taken together, the results suggest a model of complex modular control of expression of the SM-MHC gene that varies between SMC subtypes. Moreover, the studies establish the possibility of designing derivatives of the SM-MHC promoter that might be used for targeting gene expression to specific SMC subtypes in vivo.
Collapse
Affiliation(s)
- I Manabe
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, USA
| | | |
Collapse
|
43
|
Phiel CJ, Gabbeta V, Parsons LM, Rothblat D, Harvey RP, McHugh KM. Differential binding of an SRF/NK-2/MEF2 transcription factor complex in normal versus neoplastic smooth muscle tissues. J Biol Chem 2001; 276:34637-50. [PMID: 11457859 DOI: 10.1074/jbc.m105826200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The malignant potential of smooth muscle tumors correlates strongly with the disappearance of gamma-smooth muscle isoactin, a lineage-specific marker of smooth muscle development. In this paper, we identify a 36-base pair regulatory motif containing an AT-rich domain, CArG box, and a non-canonical NK-2 homeodomain-binding site that has the capacity to regulate smooth muscle-specific gene expression in cultured intestinal smooth muscle cells. Serum-response factor associates with an NK-2 transcription factor via protein-protein interactions and binds to the core CArG box element. Our studies suggest that the NK-2 transcription factor that associates with serum-response factor during smooth muscle differentiation is Nkx2-3. Myocyte-specific enhancer factor 2 binding to this regulatory complex was also observed but limited to uterine smooth muscle tissues. Smooth muscle neoplasms displayed altered transcription factor binding when compared with normal myometrium. Differential nuclear accessibility of serum-response factor protein during smooth muscle differentiation and neoplastic transformation was also observed. Thus, we have identified a unique regulatory complex whose differential binding properties and nuclear accessibility are associated with modulating gamma-smooth muscle isoactin-specific gene expression in both normal and neoplastic tissues.
Collapse
Affiliation(s)
- C J Phiel
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | |
Collapse
|
44
|
Reusch HP, Zimmermann S, Schaefer M, Paul M, Moelling K. Regulation of Raf by Akt controls growth and differentiation in vascular smooth muscle cells. J Biol Chem 2001; 276:33630-7. [PMID: 11443134 DOI: 10.1074/jbc.m105322200] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The stimulation of platelet-derived growth factor (PDGF) receptors shifts vascular smooth muscle (VSM) cells toward a more proliferative phenotype. Thrombin activates the same signaling cascades in VSM cells, namely the Ras/Raf/MEK/ERK and the phosphatidylinositol 3-kinase (PI 3-kinase)/Akt pathways. Nonetheless, thrombin was not mitogenic, but rather increased the expression of the smooth muscle-specific myosin heavy chain (SM-MHC) indicative of an in vitro re-differentiation of VSM cells. A more detailed analysis of the temporal pattern and relative signal intensities revealed marked differences. The strong and biphasic phosphorylation of ERK1/2 in response to thrombin correlated with its ability to increase the activity of the SM-MHC promoter whereas Akt was only partially and transiently phosphorylated. By contrast, PDGF, a potent mitogen in VSM cells, induced a short-lived ERK1/2 phosphorylation but a complete and sustained phosphorylation of Akt. The phosphorylated form of Akt physically interacted with Raf. Moreover, Akt phosphorylated Raf at Ser(259), resulting in a reduced Raf kinase activity and a termination of MEK and ERK1/2 phosphorylation. Disruption of the PI 3-kinase signaling prevented the PDGF-induced Akt and Raf-Ser(259) phosphorylation. Under these conditions, PDGF elicited a more sustained MEK and ERK phosphorylation and increased SM-MHC promoter activity. Consistently, in cells that express dominant negative Akt, PDGF increased SM-MHC promoter activity. Furthermore, expression of constitutively active Akt blocked the thrombin-stimulated SM-MHC promoter activity. Thus, we present evidence that the balance and cross-regulation between the PI 3-kinase/Akt and Ras/Raf/MEK signaling cascades determine the temporal pattern of ERK1/2 phosphorylation and may thereby guide the phenotypic modulation of vascular smooth muscle cells.
Collapse
Affiliation(s)
- H P Reusch
- Institut für Klinische Pharmakologie und Toxikologie, Freie Universität Berlin, Garystr. 5, 14195 Berlin, Germany.
| | | | | | | | | |
Collapse
|
45
|
Reusch HP, Schaefer M, Plum C, Schultz G, Paul M. Gbeta gamma mediate differentiation of vascular smooth muscle cells. J Biol Chem 2001; 276:19540-7. [PMID: 11279222 DOI: 10.1074/jbc.m101963200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Proliferation and subsequent dedifferentiation of vascular smooth muscle (VSM) cells contribute to the pathogenesis of atherosclerosis and postangioplastic restenosis. The dedifferentiation of VSM cells in vivo or in cell culture is characterized by a loss of contractile proteins such as smooth muscle-specific alpha-actin and myosin heavy chain (SM-MHC). Serum increased the expression of contractile proteins in neonatal rat VSM cells, indicating a redifferentiation process. RNase protection assays defined thrombin as a serum component that increases the abundance of SM-MHC transcripts. Additionally, serum and thrombin transiently elevated cytosolic Ca(2+) concentrations, led to a biphasic extracellular signal-regulated kinase (ERK) phosphorylation, up-regulated a transfected SM-MHC promoter construct, and induced expression of the contractile proteins SM-MHC and alpha-actin. Pertussis toxin, N17-Ras/Raf, and PD98059 prevented both the serum- and thrombin-induced second phase ERK phosphorylation and SM-MHC promoter activation. Constitutively active Galpha(q), Galpha(i), Galpha(12), and Galpha(13) failed to up-regulate SM-MHC transcription, whereas Gbetagamma concentration-dependently increased the SM-MHC promoter activity. Furthermore, the Gbetagamma scavenger beta-adrenergic receptor kinase 1 C-terminal peptide abolished the serum-mediated differentiation. We conclude that receptor-mediated differentiation of VSM cells requires Gbetagamma and an intact Ras/Raf/MEK/ERK signaling.
Collapse
MESH Headings
- Actins/biosynthesis
- Animals
- Animals, Newborn
- Calcium/metabolism
- Cell Differentiation
- Cell Division
- Cells, Cultured
- Cytosol/metabolism
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Flavonoids/pharmacology
- GTP-Binding Protein beta Subunits
- GTP-Binding Protein gamma Subunits
- Heterotrimeric GTP-Binding Proteins/metabolism
- Heterotrimeric GTP-Binding Proteins/physiology
- Immunoblotting
- Microscopy, Fluorescence
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- Myosin Heavy Chains/biosynthesis
- Pertussis Toxin
- Phosphorylation
- Promoter Regions, Genetic
- RNA, Messenger/metabolism
- Rats
- Ribonucleases/metabolism
- Signal Transduction
- Thrombin/metabolism
- Time Factors
- Transcription, Genetic
- Transfection
- Up-Regulation
- Virulence Factors, Bordetella/pharmacology
Collapse
Affiliation(s)
- H P Reusch
- Institute of Clinical Pharmacology and Toxicology and the Institute of Pharmacology, Benjamin Franklin Medical Center, Freie Universität Berlin, Berlin 14195, Germany
| | | | | | | | | |
Collapse
|
46
|
Arafat HA, Kim GS, DiSanto ME, Wein AJ, Chacko S. Heterogeneity of bladder myocytes in vitro: modulation of myosin isoform expression. Tissue Cell 2001; 33:219-32. [PMID: 11469535 DOI: 10.1054/tice.2001.0171] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We studied the expression of myosin heavy chain isoforms differing at the N-terminal (SM-A, SM-B) and the C-terminal (SM1, SM2) regions and non-muscle myosin heavy chain II-A and II-B (NMMHC II-A and B) in newborn and adult rabbit bladder smooth muscle cells (SMCs) and in cultures of enzymatically dissociated neonatal detrusor. RT-PCR analyses revealed that 94.5+/-3.27% of MHC transcripts of the adult bladder SMCs contained the 21-nucleotide insert (SM-B) compared with 83.8+/-3.2% in the newborn bladder, with the remainder of the mRNA being non-inserted (SM-A). In 3, 7, and 10 days of primary culture (proliferating, confluent, and post-confluent, respectively) and up to 4 subculture passages, bladder myocytes expressed predominantly SM-A. Immunofluorescence microscopy revealed heterogeneity in cultured myocytes, i.e. SM-B positive cells coexisting with negatively stained cells. In adult bladder, the C-terminal isoforms SM1 and SM2 represented, 43.1+/-4.3% and 56.89 + 4.3% of the mRNA, respectively, while newborn bladders expressed 72.5+/-7% SM1 and 27.5+/-7% SM2. Upon culturing, cells predominantly expressed SM1 at both the mRNA and protein levels. NMMHC II-A was expressed by both adult and newborn bladders and in culture, whereas NMMHC II-B was expressed at low levels only in newborn bladders, but upregulated in culture. These data indicate that bladder myocytes in vitro undergo modulation with relative overexpression of SM-A and SM1 and upregulation of NMMHC II-B. Information on the mechanisms responsible for this modulation in vitro might provide an understanding of the nature of altered myosin isoform expression associated with smooth muscle dysfunction in certain bladder diseases.
Collapse
Affiliation(s)
- H A Arafat
- Division of Urology, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | |
Collapse
|
47
|
Ogra Y, Suzuki K, Gong P, Otsuka F, Koizumi S. Negative regulatory role of Sp1 in metal responsive element-mediated transcriptional activation. J Biol Chem 2001; 276:16534-9. [PMID: 11279094 DOI: 10.1074/jbc.m100570200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transcription of mammalian metallothionein (MT) genes is activated by heavy metals via multiple copies of a cis-acting DNA element, the metal-responsive element (MRE). Our previous studies have shown that certain MREs of the human MT-IIA gene (MREb, MREc, MREd, and MREf) are less active than the others (MREa, MREe, and MREg). Gel shift analysis of HeLa cell nuclear proteins revealed that whereas the active MREs strongly bind the transcription factor MTF-1 essential for metal regulation, the less active MREs bind another distinct protein, MREb-BF. This protein recognizes the GC-rich region of MREb rather than the MRE core required for MTF-1 binding. All the MREs recognized by MREb-BF contain the CGCCC and/or CACCC motif, suggesting that the MREb-BF.MRE complex contains Sp1 or related proteins. Supershift analysis using antibodies against Sp1 family proteins as well as gel shift analysis using the recombinant Sp1 demonstrated that Sp1 represents the majority of MREb-BF activity. An MREb mutant with reduced affinity to Sp1 mediated zinc-inducible transcription much more actively than the wild-type MREb. Furthermore, when placed in the native promoter, this mutant MREb raised the overall promoter activity. These results strongly suggest that Sp1 acts as a negative regulator of transcription mediated by specific MREs.
Collapse
Affiliation(s)
- Y Ogra
- Divisions of Hazard Assessment and Health Effects Research, National Institute of Industrial Health, 6-21-1, Nagao, Tama-ku, Kawasaki 214-8585, Japan
| | | | | | | | | |
Collapse
|
48
|
Herring BP, Kriegel AM, Hoggatt AM. Identification of Barx2b, a serum response factor-associated homeodomain protein. J Biol Chem 2001; 276:14482-9. [PMID: 11278942 DOI: 10.1074/jbc.m011585200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CC(A/T)(6)GG or serum response elements represent a common regulatory motif important for regulating the expression of many smooth muscle-specific genes. They are multifunctional elements that bind serum response factor (SRF) and are important for serum induction of genes, expression of muscle-specific genes, and differentiation of vascular smooth muscle cells. In the current study, a yeast two-hybrid screen was used to identify proteins from mouse intestine that interact with SRF. A novel homeodomain-containing transcription factor, called Barx2b, was identified that specifically interacts with SRF and promotes the DNA binding activity of SRF. Northern blotting, RNase protection analysis, and Western blotting revealed that Barx2b mRNA and protein are expressed in several smooth muscle-containing tissues, as well as in skeletal muscle and brain. In vitro binding studies using bacterial fusion proteins revealed that the DNA-binding domain of SRF interacts with a region of Barx2b located amino-terminal of the homeobox domain. The results of these studies support the hypothesis that interaction of SRF with different homeodomain-containing proteins may play a critical role in determining the cell-specific functions of SRF.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Blotting, Northern
- Blotting, Western
- Brain/metabolism
- Cell Differentiation
- Cell Line
- DNA/metabolism
- DNA, Complementary/metabolism
- DNA-Binding Proteins/metabolism
- Electrophoresis, Polyacrylamide Gel
- Gene Library
- Genes, Reporter
- Homeodomain Proteins/biosynthesis
- Homeodomain Proteins/chemistry
- Homeodomain Proteins/metabolism
- Intestinal Mucosa/metabolism
- Mice
- Models, Genetic
- Molecular Sequence Data
- Muscle, Skeletal/metabolism
- Muscle, Smooth/metabolism
- Nuclear Proteins/metabolism
- Protein Binding
- RNA, Messenger/metabolism
- Recombinant Fusion Proteins/metabolism
- Recombinant Proteins/metabolism
- Ribonucleases/metabolism
- Sequence Homology, Amino Acid
- Serum Response Factor
- Time Factors
- Tissue Distribution
- Two-Hybrid System Techniques
Collapse
Affiliation(s)
- B P Herring
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis 46202-5120, USA.
| | | | | |
Collapse
|
49
|
Manabe I, Owens GK. CArG elements control smooth muscle subtype-specific expression of smooth muscle myosin in vivo. J Clin Invest 2001; 107:823-34. [PMID: 11285301 PMCID: PMC199571 DOI: 10.1172/jci11385] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Expression of smooth muscle myosin heavy chain (SM-MHC) is tightly controlled depending on the differentiated state of smooth muscle cells (SMCs). To better understand the mechanisms that regulate transcription of the SM-MHC gene in vivo, we tested the function of several conserved CArG elements contained within the -4200 to +11,600 region of this gene that we had previously shown to drive SMC-specific expression in transgenic mice. CArG1 in the 5'-flanking sequence was required for all SMCs, while CArG2 and a novel intronic CArG element were differentially required in SMC subtypes. Of particular note, mutation of the intronic CArG selectively abolished expression in large arteries. A promoter construct containing three repeats of a conserved 227-bp intronic CArG-containing region was sufficient to direct transcription in vascular SMCs in transgenic mice, although this construct was also expressed in skeletal and cardiac muscle. These results support a model in which transcriptional regulation of SM-MHC is controlled by multiple positive and negative modular control regions that differ between SMCs and non-SMCs and among SMC subtypes. We also demonstrated that the CArG elements of the endogenous SM-MHC gene were bound by SRF in chromatin.
Collapse
Affiliation(s)
- I Manabe
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, USA
| | | |
Collapse
|
50
|
Schön U, Seifarth W, Baust C, Hohenadl C, Erfle V, Leib-Mösch C. Cell type-specific expression and promoter activity of human endogenous retroviral long terminal repeats. Virology 2001; 279:280-91. [PMID: 11145909 DOI: 10.1006/viro.2000.0712] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Evolution over millions of years has adapted several thousand copies of retrovirus-like elements and over 10 times as many solitary long terminal repeats (LTRs) to their present location in the human genome. Transcription of these human endogenous retroviruses (HERVs) has been detected in various cells and tissues, and in some cases their transcriptional control elements have been recruited by cellular genes. We used a retroviral pol-specific expression array to obtain a HERV transcription profile in a variety of human cells such as epidermal keratinocytes, liver cells, kidney cells, pancreatic cells, lymphocytes, and lung fibroblasts. This rapid screening test revealed a distinct HERV pol-expression pattern in each cell type tested so far. About 40 different U3/R regulatory sequences from the HERV-H and HERV-W families were then amplified from actively transcribed 3'HERV LTRs of various cell lines and tissues. Their promoter activities were compared with LTR sequences of other known HERV families in 12 human cell lines using a transient luciferase reporter system. Expression of the isolated HERV LTRs varied significantly in these cell lines, in some cases showing strict cell type specificity. These results suggest that endogenous retroviral LTRs may be a valuable source of transcriptional regulatory elements for the construction of targeted retroviral expression vectors.
Collapse
Affiliation(s)
- U Schön
- Institute of Molecular Virology, Oberschleissheim, D-85764, Germany.
| | | | | | | | | | | |
Collapse
|