1
|
Thota LNR, Chignalia AZ. The role of the glypican and syndecan families of heparan sulfate proteoglycans in cardiovascular function and disease. Am J Physiol Cell Physiol 2022; 323:C1052-C1060. [PMID: 35993518 DOI: 10.1152/ajpcell.00018.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) are proteoglycans formed by a core protein to which one or multiple heparan sulfate chains are covalently bound. They are ubiquitously expressed in cellular surfaces and can be found in the extracellular matrix and secretory vesicles. The cellular effects of HSPGs comprehend multiple functionalities that include 1) the interaction with other membrane surface proteins to act as a substrate for cellular migration, 2) acting as a binding site for circulating molecules, 3) to have a receptor role for proteases, 4) to act as a coreceptor that can provide finetuning of growth factor receptor activity threshold, and 5) to activate intracellular signaling pathways (Sarrazin S, Lamanna WC, Esko JD. Cold Spring Harb Perspect Biol 3: a004952, 2011). Among the different families of HSPGs, the syndecan and glypican families of HSPGs have gained increased attention in relation to their effects on cardiovascular cells and potential role in disease progression. In this review, we will summarize the effects of syndecan and glypican homologs on the different cardiovascular cell types and discuss their contribution to common processes found in cardiovascular diseases (inflammation, hypertrophy, and vascular remodeling) as well as their potential role in the development and progression of specific diseases including hypertension, heart failure, and atherosclerosis.
Collapse
Affiliation(s)
| | - Andreia Zago Chignalia
- Department of Anesthesiology, College of Medicine-Tucson, University of Arizona, Tucson, Arizona.,Department of Physiology, College of Medicine-Tucson, University of Arizona, Tucson, Arizona.,Department of Pharmacology and Toxicology, College of Pharmacy-Tucson, University of Arizona, Tucson, Arizona.,Sarver Heart Center, College of Medicine-Tucson, University of Arizona, Tucson, Arizona
| |
Collapse
|
2
|
Lord MS, Tang F, Rnjak-Kovacina J, Smith JGW, Melrose J, Whitelock JM. The multifaceted roles of perlecan in fibrosis. Matrix Biol 2018; 68-69:150-166. [PMID: 29475023 DOI: 10.1016/j.matbio.2018.02.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/11/2022]
Abstract
Perlecan, or heparan sulfate proteoglycan 2 (HSPG2), is a ubiquitous heparan sulfate proteoglycan that has major roles in tissue and organ development and wound healing by orchestrating the binding and signaling of mitogens and morphogens to cells in a temporal and dynamic fashion. In this review, its roles in fibrosis are reviewed by drawing upon evidence from tissue and organ systems that undergo fibrosis as a result of an uncontrolled response to either inflammation or traumatic cellular injury leading to an over production of a collagen-rich extracellular matrix. This review focuses on examples of fibrosis that occurs in lung, liver, kidney, skin, kidney, neural tissues and blood vessels and its link to the expression of perlecan in that particular organ system.
Collapse
Affiliation(s)
- Megan S Lord
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia.
| | - Fengying Tang
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia
| | | | - James G W Smith
- University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - James Melrose
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; Sydney Medical School, Northern, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, UNSW Sydney, NSW 2052, Australia
| |
Collapse
|
3
|
Tran-Lundmark K, Tannenberg P, Rauch BH, Ekstrand J, Tran PK, Hedin U, Kinsella MG. Perlecan Heparan Sulfate Is Required for the Inhibition of Smooth Muscle Cell Proliferation by All-trans-Retinoic Acid. J Cell Physiol 2015; 230:482-7. [PMID: 25078760 DOI: 10.1002/jcp.24731] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 07/25/2014] [Indexed: 12/13/2022]
Abstract
Smooth muscle cell (SMC) proliferation is a key process in stabilization of atherosclerotic plaques, and during restenosis after interventions. A clearer understanding of SMC growth regulation is therefore needed to design specific anti-proliferative therapies. Retinoic acid has been shown to inhibit proliferation of SMCs both in vitro and in vivo and to affect the expression of extracellular matrix molecules. To explore the mechanisms behind the growth inhibitory activity of retinoic acid, we hypothesized that retinoids may induce the expression of perlecan, a large heparan sulfate proteoglycan with anti-proliferative properties. Perlecan expression and accumulation was induced in murine SMC cultures by all-trans-retinoic acid (AtRA). Moreover, the growth inhibitory effect of AtRA on wild-type cells was greatly diminished in SMCs from transgenic mice expressing heparan sulfate-deficient perlecan, indicating that the inhibition is perlecan heparan sulfate-dependent. In addition, AtRA influenced activation and phosphorylation of PTEN and Akt differently in wild-type and mutant SMCs, consistent with previous studies of perlecan-dependent SMC growth inhibition. We demonstrate that AtRA regulates perlecan expression in SMCs and that the inhibition of SMC proliferation by AtRA is, at least in part, secondary to an increased expression of perlecan and dependent upon its heparan sulfate-chains.
Collapse
Affiliation(s)
- Karin Tran-Lundmark
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Philip Tannenberg
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Bernhard H Rauch
- Institute of Pharmacology, Center of Drug Absorption and Transport, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Johan Ekstrand
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Phan-Kiet Tran
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
4
|
Zhu K, Wang H, Gul Y, Zhao Y, Wang W, Liu S, Wang M. Expression characterization and the promoter activity analysis of zebrafish hdac4. FISH PHYSIOLOGY AND BIOCHEMISTRY 2012; 38:585-593. [PMID: 21773810 DOI: 10.1007/s10695-011-9540-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 07/04/2011] [Indexed: 05/31/2023]
Abstract
Histone deacetylase 4 (HDAC4) is an important modifier enzyme for chromatin remodeling and plays an essential role in regulating gene expression. Spatio-temporal expression spectrum revealed that zebrafish hdac4 mRNA, ubiquitously distributed in various tissues, were significantly higher at 36 hpf (hours post-fertilization) and 6 dpf (days post-fertilization) than other periods. Trichostatin A (TSA) inhibited the development of zebrafish embryos and transcription of hdac4 and mef2a (myocyte enhancer factor-2A). Moreover, five vectors containing different promoter regions of hdac4 were constructed in order to analyze promoter activity. The vector containing the region from -125 to +160 exhibited maximum luciferase activity that was approximately 30.3-fold and 58.9-fold higher than the control in two kinds of cells, respectively. By comparing the luciferase activities between the region from -302 to +30 and -698 to +30, it was suggested that the region between -698 and -302 might contain mild negative regulatory elements.
Collapse
Affiliation(s)
- Kecheng Zhu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
5
|
Liu Y, Li Y, Li T, Lu H, Jia Z, Wang W, Chen P, Ma K, Zhou C. POU homeodomain protein OCT1 modulates islet 1 expression during cardiac differentiation of P19CL6 cells. Cell Mol Life Sci 2011; 68:1969-82. [PMID: 20960024 PMCID: PMC11115038 DOI: 10.1007/s00018-010-0544-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 09/02/2010] [Accepted: 09/28/2010] [Indexed: 11/30/2022]
Abstract
Islet 1 (ISL1), a marker of cardiac progenitors, plays a crucial role in cardiogenesis. However, the precise mechanism underlying the activation of its expression is not fully understood. Using the cardiac differentiation model of P19CL6 cells, we show that POU homeodomain protein, OCT1, modulates Isl1 expression in the process of cardiac differentiation. Oct1 knock-down resulted in reduction of Isl1 expression and downregulated mesodermal, cardiac-specific, and signal pathway gene expression. Additionally, the octamer motif located in the proximal region of Isl1 promoter is essential to Isl1 transcriptional activation. Mutation of this motif remarkably decreased Isl1 transcription. Although both OCT1 and OCT4 bound to this motif, it was OCT1 rather than OCT4 that modulated Isl1 expression. Furthermore, the correlation of OCT1 in regulation of Isl1 was revealed by in situ hybridization in early embryos. Collectively, our data highlight a novel role of OCT1 in the regulation of Isl1 expression.
Collapse
Affiliation(s)
- Yinan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
| | - Yanming Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
| | - Tao Li
- Department of Biology, College of Chemistry and Life Science, Zhejiang Normal University, 688 Yingbing Road, Jinhua, 321004 Zhejiang Province China
| | - Huafei Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
| | - Zhuqing Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| | - Weiping Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| | - Ping Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| | - Kangtao Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| | - Chunyan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Road, Haidian District Beijing, 100191 China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191 China
| |
Collapse
|
6
|
Xu L, Matsumoto A, Sasaki A, Harada H, Taniguchi A. Identification of a suppressor element in the amelogenin promoter. J Dent Res 2009; 89:246-51. [PMID: 20044581 DOI: 10.1177/0022034509355144] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Amelogenin expression is regulated in a cell-type- specific manner. Investigators have previously identified an enhancer element by using the 5' flanking sequence of the amelogenin promoter. However, the cell-type-specific regulation of the amelogenin gene remains poorly understood. In some genes, the first intron regulates tissue-specific expression. We hypothesized that intron 1 is important for the cell-type-specific regulation of amelogenin expression. We identified a suppressor element between -74 and -464. We also found enhancer activity in intron 1. Additionally, we found that the suppressor element in the promoter region suppressed intron 1 enhancer activity. The suppressor and the enhancers acted in an ameloblast-like cell line, but not in HeLa cells. Mutation of the Oct-1 binding sites reversed the suppressor activity, suggesting that Oct-1 sites are essential for suppression. These results suggest that Oct-1 and intron 1 may contribute to cell-type-specific amelogenin expression.
Collapse
Affiliation(s)
- L Xu
- Advanced Medical Materials Group, Biomaterials Center, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | | | | | | | | |
Collapse
|
7
|
dela Paz NG, Simeonidis S, Leo C, Rose DW, Collins T. Regulation of NF-kappaB-dependent gene expression by the POU domain transcription factor Oct-1. J Biol Chem 2007; 282:8424-34. [PMID: 17192276 DOI: 10.1074/jbc.m606923200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Maintenance of the cells of the vessel wall in a quiescent state is an important aspect of normal vascular physiology. Transcriptional repressors are widely believed to regulate this process, yet the exact factors involved and the mechanism of repression are not known. Here, we report that the POU domain transcription factor Oct-1 represses the expression of E-selectin and vascular cell adhesion molecule (VCAM-1), two cytokine-inducible, NF-kappaB-dependent endothelial-leukocyte adhesion molecules that participate in the leukocyte recruitment phase of the inflammatory response. Co-transfection and microinjection studies demonstrate that Oct-1 blocks tumor necrosis factor alpha-stimulated E-selectin and VCAM-1 expression. Gene expression arrays indicate that control of tumor necrosis factor alpha-induced, NF-kappaB-dependent gene expression by Oct-1 is promoter-specific. A DNA-binding mutant of Oct-1 represses NF-kappaB-dependent reporter gene expression. Biochemically, Oct-1 interacts with p65, suggesting that Oct-1 is involved in the regulation of NF-kappaB transactivation function. NF-kappaB-dependent gene expression is more pronounced in Oct-1-deficient than in wild-type murine embryonic fibroblasts, and reintroduction of human Oct-1 abolishes these differences. Finally, the cytokine interleukin-6 induces Oct-1 gene expression, providing a biologically relevant means by which NF-kappaB-dependent gene expression can be selectively reverted by Oct-1 to quiescent levels.
Collapse
Affiliation(s)
- Nathaniel G dela Paz
- Molecular Pathology Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California 92093-0673, and Department of Pathology, Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
8
|
Whitelock JM, Iozzo RV. Heparan Sulfate: A Complex Polymer Charged with Biological Activity. Chem Rev 2005; 105:2745-64. [PMID: 16011323 DOI: 10.1021/cr010213m] [Citation(s) in RCA: 321] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- John M Whitelock
- Graduate School of Biomedical Engineering, University of New South Wales, Kensington, Sydney, New South Wales 2052, Australia.
| | | |
Collapse
|
9
|
Garl PJ, Wenzlau JM, Walker HA, Whitelock JM, Costell M, Weiser-Evans MCM. Perlecan-induced suppression of smooth muscle cell proliferation is mediated through increased activity of the tumor suppressor PTEN. Circ Res 2003; 94:175-83. [PMID: 14656929 DOI: 10.1161/01.res.0000109791.69181.b6] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We were interested in the elucidation of the interaction between the heparan sulfate proteoglycan, perlecan, and PTEN in the regulation of vascular smooth muscle cell (SMC) growth. We verified serum-stimulated DNA synthesis, and Akt and FAK phosphorylation were significantly reduced in SMCs overexpressing wild-type PTEN. Our previous studies showed perlecan is a potent inhibitor of serum-stimulated SMC growth. We report in the present study, compared with SMCs plated on fibronectin, serum-stimulated SMCs plated on perlecan exhibited increased PTEN activity, decreased FAK and Akt activities, and high levels of p27, consistent with SMC growth arrest. Adenoviral-mediated overexpression of constitutively active Akt reversed perlecan-induced SMC growth arrest while morpholino antisense-mediated loss of endogenous PTEN resulted in increased growth and phosphorylation of FAK and Akt of SMCs on perlecan. Immunohistochemical and Western analyses of balloon-injured rat carotid artery tissues showed a transient increase in phosphoPTEN (inactive) after injury, correlating to high rates of neointimal cell replication; phosphoPTEN was largely limited to actively replicating SMCs. Similarly, in the developing rat aorta, we found increased PTEN activity associated with increased perlecan deposition and decreased SMC replication rates. However, significantly decreased PTEN activity was detected in aortas of perlecan-deficient mouse embryos, consistent with SMC hyperplasia observed in these animals, compared with E17.5 heterozygous controls that produce abundant amounts of perlecan at this developmental time point. Our data show PTEN is a potent endogenously produced inhibitor of SMC growth and increased PTEN activity mediates perlecan-induced suppression of SMC proliferation.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/embryology
- Basement Membrane/physiology
- Carotid Artery Injuries/pathology
- Catheterization/adverse effects
- Cell Division/drug effects
- Cells, Cultured
- Culture Media, Serum-Free
- DNA Replication/drug effects
- Fibronectins/pharmacology
- Focal Adhesion Kinase 1
- Focal Adhesion Protein-Tyrosine Kinases
- Glycosaminoglycans/physiology
- Heparan Sulfate Proteoglycans/deficiency
- Heparan Sulfate Proteoglycans/pharmacology
- Heparan Sulfate Proteoglycans/physiology
- Heparitin Sulfate/physiology
- Male
- Mice
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Oligonucleotides, Antisense/pharmacology
- Phosphorylation
- Protein Processing, Post-Translational
- Protein Serine-Threonine Kinases
- Protein-Tyrosine Kinases/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- Rats
- Rats, Sprague-Dawley
- Recombinant Fusion Proteins/physiology
Collapse
Affiliation(s)
- Pamela J Garl
- Department of Pediatrics, University of Colorado Health Sciences Center, Denver, Colo 80262, USA
| | | | | | | | | | | |
Collapse
|
10
|
Wedgwood S, Black SM. Induction of apoptosis in fetal pulmonary arterial smooth muscle cells by a combined superoxide dismutase/catalase mimetic. Am J Physiol Lung Cell Mol Physiol 2003; 285:L305-12. [PMID: 12665466 DOI: 10.1152/ajplung.00382.2002] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Reactive oxygen species (ROS) such as superoxide and hydrogen peroxide are known to play an important role in the proliferation and viability of vascular smooth muscle cells. In this study, we determined the effects of increased superoxide dismutase and catalase activity on fetal pulmonary arterial smooth muscle cell (FPASMC) proliferation and viability using EUK-134, a superoxide dismutase/catalase mimetic. Treatment of FPASMC with EUK-134 or with a combination of superoxide dismutase and catalase enzymes decreased superoxide and hydrogen peroxide levels as detected by the fluorescent dyes dihydroethidium and dichlorodihydrofluorescein diacetate, respectively. EUK-134 (5 microM) attenuated serum-induced FPASMC proliferation, whereas 50 microM EUK-134 decreased the number of viable cells, suggesting cell death. Conversely, combined superoxide dismutase and catalase enzyme activity equivalent to 50 microM EUK-134 prevented proliferation but did not reduce the number of viable FPASMC. The loss of mitochondrial membrane potential after 18 h, an increase in caspase-9 and caspase-3 activity after 24 h, and the subsequent appearance of TdT-mediated dUTP nick end labeling-positive nuclei were detected in FPASMC after treatment with 50 microM EUK-134. This indicates an induction of programmed rather than necrotic cell death and suggests that prolonged removal of ROS is required to stimulate apoptosis. Compounds such as EUK-134 may, therefore, prove more effective than enzymic antioxidants over longer periods, especially when the aim is to decrease the number of smooth muscle cells in diseases resulting from excessive muscularization.
Collapse
Affiliation(s)
- Stephen Wedgwood
- Department of Pediatrics, Northwestern University Medical School, Chicago, IL 60611-3008, USA
| | | |
Collapse
|
11
|
Walker HA, Whitelock JM, Garl PJ, Nemenoff RA, Stenmark KR, Weiser-Evans MCM. Perlecan up-regulation of FRNK suppresses smooth muscle cell proliferation via inhibition of FAK signaling. Mol Biol Cell 2003; 14:1941-52. [PMID: 12802066 PMCID: PMC165088 DOI: 10.1091/mbc.e02-08-0508] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2002] [Revised: 12/17/2002] [Accepted: 12/27/2002] [Indexed: 12/25/2022] Open
Abstract
We previously reported that fully assembled basement membranes are nonpermissive to smooth muscle cell (SMC) replication and that perlecan (PN), a basement membrane heparan sulfate proteoglycan, is a dominant effector of this response. We report here that SMC adhesion to basement membranes, and perlecan in particular, up-regulate the expression of focal adhesion kinase-related nonkinase (FRNK), a SMC-specific endogenous inhibitor of FAK, which subsequently suppresses FAK-mediated, ERK1/2-dependent growth signals. Up-regulation of FRNK by perlecan is actively and continuously regulated. Relative to the matrix proteins studied, the effects are unique to perlecan, because plating of SMCs on several other basement membrane proteins is associated with low levels of FRNK and corresponding high levels of FAK and ERK1/2 phosphorylation and SMC growth. Perlecan supports SMC adhesion, although there is reduced cell spreading compared with fibronectin (FN), laminin (LN), or collagen type IV (IV). Despite the reduction in cell spreading, we report that perlecan-induced up-regulation of FRNK is independent of cell shape changes. Growth inhibition by perlecan was rescued by overexpressing a constitutively active FAK construct, but overexpressing kinase-inactivated mutant FAK or FRNK attenuated fibronectin-stimulated growth. These data indicate that perlecan functions as an endogenously produced inhibitor of SMC growth at least in part through the active regulation of FRNK expression. FRNK, in turn, may control SMC growth by downregulating FAK-dependent signaling events.
Collapse
Affiliation(s)
- Heather A Walker
- Department of Pediatrics, Renal Division, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | | | | | | | |
Collapse
|
12
|
Kinsella MG, Tran PK, Weiser-Evans MCM, Reidy M, Majack RA, Wight TN. Changes in perlecan expression during vascular injury: role in the inhibition of smooth muscle cell proliferation in the late lesion. Arterioscler Thromb Vasc Biol 2003; 23:608-14. [PMID: 12615671 DOI: 10.1161/01.atv.0000063109.94810.ee] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular smooth muscle cells (SMCs), activated by growth factors after arterial injury, migrate and proliferate to expand the intima of the blood vessel. During intimal expansion, proliferation is suppressed and an increasingly large proportion of the neointimal mass is composed of newly synthesized extracellular matrix (ECM). We sough to determine whether the ECM heparan sulfate proteoglycan (HSPG) perlecan, which inhibits SMC proliferation in vitro, also accumulates and limits SMC proliferation during neointimal expansion. METHODS AND RESULTS Perlecan expression and accumulation were analyzed by immunohistochemistry and in situ hybridization during neointima formation after balloon catheter injury to the rat carotid artery. Perlecan expression was low in uninjured vessels and up to 7 days after injury, during maximal SMC proliferation. By 14 days after injury, perlecan was dramatically increased, and immunostaining remained heavy throughout the advanced lesion, 35 to 42 days after injury. Finally, explants of intimal tissue from 35- to 42-day neointimal lesions were digested with glycosaminoglycanases to determine whether endogenous HSPGs inhibit intimal SMC proliferation. SMCs within HS-depleted, but not chondroitinase ABC-treated or mock-incubated, explants were found to proliferate in response to platelet-derived growth factor BB. CONCLUSIONS HSPGs, such as perlecan, may inhibit the proliferative response of SMCs after vascular injury.
Collapse
Affiliation(s)
- Michael G Kinsella
- Department of Pathology, University of Washington, and the Hope Heart Institute, Seattle, Wash 98104, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Markmann A, Rauterberg J, Vischer P, Robenek H, Echtermeyer F, Will H, Seidler DG, Young MF, Kresse H. Expression of transcription factors and matrix genes in response to serum stimulus in vascular smooth muscle cells. Eur J Cell Biol 2003; 82:119-29. [PMID: 12691261 DOI: 10.1078/0171-9335-00309] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During atherogenesis vascular smooth muscle cells are converted from a contractile into a synthetic phenotype characterized by enhanced matrix production. The transcription factors Gax and GATA-6 are considered negative, and Oct-1 positive regulators of the synthetic phenotype. Since the phenotype transition can be induced by culturing the cells with serum, we followed the expression of Gax, GATA-6 and Oct-1, integrins and matrix genes in quiescent porcine vascular smooth muscle cells after serum application. Comparisons were made between enzymatically released primary smooth muscle cells and cells grown out from explants of the medial layer of porcine aorta. The serum-mediated down-regulation of Gax was more intense than that of GATA-6, and stronger in explant-derived than in primary cells. Serum was without influence on the expression of Oct-1. Changes in the expression of the transcription factors preceded the induction of integrin alpha2 and the down-regulation of decorin, while mRNAs for laminin beta1 and osteopontin rose immediately after serum stimulation. Primary cells reacted more rapidly than explant cells with respect to changes in laminin isoforms. Studies with a Gax-expressing adenovirus indicated that among all the gene products tested only the expression of integrin alpha2 responded to Gax induction. Thus, our data show that i) Gax should be considered a transcription factor being directly responsible for only few aspects of the phenotypic conversion of smooth muscle cells and that ii) explant cells may represent a subpopulation of smooth muscle cells, which differ from the total population of smooth muscle cells, as obtained in primary culture, in their response to serum stimuli.
Collapse
Affiliation(s)
- André Markmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster/Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Mongiat M, Sweeney SM, San Antonio JD, Fu J, Iozzo RV. Endorepellin, a novel inhibitor of angiogenesis derived from the C terminus of perlecan. J Biol Chem 2003; 278:4238-49. [PMID: 12435733 DOI: 10.1074/jbc.m210445200] [Citation(s) in RCA: 262] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Perlecan, a ubiquitous basement membrane heparan sulfate proteoglycan, plays key roles in blood vessel growth and structural integrity. We discovered that the C terminus of perlecan potently inhibited four aspects of angiogenesis: endothelial cell migration, collagen-induced endothelial tube morphogenesis, and blood vessel growth in the chorioallantoic membrane and in Matrigel plug assays. The C terminus of perlecan was active at nanomolar concentrations and blocked endothelial cell adhesion to fibronectin and type I collagen, without directly binding to either protein; henceforth we have named it "endorepellin." We also found that endothelial cells possess a significant number of high affinity (K(d) of 11 nm) binding sites for endorepellin and that endorepellin binds endostatin and counteracts its anti-angiogenic effects. Thus, endorepellin represents a novel anti-angiogenic product, which may retard tumor neovascularization and hence tumor growth in vivo.
Collapse
Affiliation(s)
- Maurizio Mongiat
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | |
Collapse
|
15
|
Delmolino LM, Stearns NA, Castellot JJ. COP-1, a member of the CCN family, is a heparin-induced growth arrest specific gene in vascular smooth muscle cells. J Cell Physiol 2001; 188:45-55. [PMID: 11382921 DOI: 10.1002/jcp.1100] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Vascular smooth muscle cell (VSMC) hyperplasia is responsible for the failure of 15-30% of vascular surgical procedures such as coronary artery bypass grafts and angioplasties. We and others have shown that heparin suppresses VSMC proliferation in vivo and in cell culture. We hypothesize that heparin inhibits VSMC proliferation by binding to cell surface receptors, resulting in selective modulation of mitogenic signal transduction pathways and altered transcription of a specific subset of growth regulatory genes. To test this idea, we used subtractive hybridization to identify differentially expressed mRNAs in heparin-treated and untreated VSMC. We identified a heparin induced mRNA identical to Cop-1, a member of the CCN family of proteins which are secreted, cysteine-rich modular proteins involved in growth regulation and migration. Cop-1 from smooth muscle cells appears to have a different expression pattern and possibly different functions than Cop-1 from other cells. Cop-1 mRNA is expressed at high levels in quiescent VSMC and at low levels in proliferating VSMC, an expression pattern highly characteristic of growth arrest specific genes. Cop-1 mRNA is expressed at high levels in heparin treated VSMC and COP-1 protein is secreted into culture medium. In tissues, Cop-1 expression is observed in the uninjured rat aorta suggesting a possible role for Cop-1 in vivo. We found PDGF, but not EGF, inhibits the expression of Cop-1 in VSMC. Neither TGF-beta nor interferon-beta, two inhibitors of VSMC proliferation, were able to induce Cop-1 expression. In addition, heparin does not induce Cop-1 mRNA in endothelial cells and VSMC resistant to the antiproliferative effect of heparin. Conditioned medium from cells over-expressing COP-1 protein inhibits VSMC proliferation in culture. Together, our data indicate that COP-1 may play a role in the antiproliferative mechanism of action of heparin.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Aorta
- Base Sequence
- Blotting, Northern
- CCN Intercellular Signaling Proteins
- Cell Division/physiology
- Cells, Cultured
- Culture Media, Serum-Free
- Endothelium, Vascular/cytology
- Gene Library
- Growth Inhibitors/chemistry
- Growth Inhibitors/genetics
- Growth Inhibitors/metabolism
- Heparin/pharmacology
- Male
- Molecular Sequence Data
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Protein Structure, Tertiary
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Repressor Proteins/chemistry
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Sequence Alignment
Collapse
Affiliation(s)
- L M Delmolino
- Department of Pathology, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
16
|
Lundmark K, Tran PK, Kinsella MG, Clowes AW, Wight TN, Hedin U. Perlecan inhibits smooth muscle cell adhesion to fibronectin: role of heparan sulfate. J Cell Physiol 2001; 188:67-74. [PMID: 11382923 DOI: 10.1002/jcp.1094] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Smooth muscle cell migration, proliferation, and deposition of extracellular matrix are key events in atherogenesis and restenosis development. To explore the mechanisms that regulate smooth muscle cell function, we have investigated whether perlecan, a basement membrane heparan sulfate proteoglycan, modulates interaction between smooth muscle cells and other matrix components. A combined substrate of fibronectin and perlecan showed a reduced adhesion of rat aortic smooth muscle cells by 70-90% in comparison to fibronectin alone. In contrast, perlecan did not interfere with cell adhesion to laminin. Heparinase treated perlecan lost 60% of its anti-adhesive effect. Furthermore, heparan sulfate as well as heparin reduced smooth muscle cell adhesion when combined with fibronectin whereas neither hyaluronan nor chondroitin sulfate had any anti-adhesive effects. Addition of heparin as a second coating to a preformed fibronectin matrix did not affect cell adhesion. Cell adhesion to the 105- and 120 kDa cell-binding fragments of fibronectin, lacking the main heparin-binding domains, was also inhibited by heparin. In addition, co-coating of fibronectin and (3)H-heparin showed that heparin was not even incorporated in the substrate. Morphologically, smooth muscle cells adhering to a substrate prepared by co-coating of fibronectin and perlecan or heparin were small, rounded, lacked focal contacts, and showed poorly developed stress fibers of actin. The results show that the heparan sulfate chains of perlecan lead to altered interactions between smooth muscle cells and fibronectin, possibly due to conformational changes in the fibronectin molecule. Such interactions may influence smooth muscle cell function in atherogenesis and vascular repair processes.
Collapse
Affiliation(s)
- K Lundmark
- Department of Surgical Sciences, Division of Vascular Surgery, Karolinska Hospital, SE-171, 76 Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
17
|
Mericskay M, Parlakian A, Porteu A, Dandré F, Bonnet J, Paulin D, Li Z. An overlapping CArG/octamer element is required for regulation of desmin gene transcription in arterial smooth muscle cells. Dev Biol 2000; 226:192-208. [PMID: 11023680 DOI: 10.1006/dbio.2000.9865] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The desmin gene encodes an intermediate filament protein that is present in skeletal, cardiac, and smooth muscle cells. This study shows that the 4-kb upstream region of the murine desmin promoter directs expression of a lacZ reporter gene throughout the heart from E7.5 and in skeletal muscle and vascular smooth muscle cells from E9. 5. The distal fragment (-4005/-2495) is active in arterial smooth muscle cells but not in venous smooth muscle cells or in the heart in vivo. It contains a CArG/octamer overlapping element (designated CArG4) that can bind the serum response factor (SRF) and an Oct-like factor. The desmin distal fragment can replace a SM22alpha regulatory region (-445/-126) that contains two CArG boxes, to cis-activate a minimal (-125/+65) SM22alpha promoter fragment in arterial smooth muscle cells of transgenic embryos. lacZ expression was abolished when mutations were introduced into the desmin CArG4 element that abolished the binding of SRF and/or Oct-like factor. These data suggest that a new type of combined CArG/octamer element plays a prominent role in the regulation of the desmin gene in arterial smooth muscle cells, and SRF and Oct-like factor could cooperate to drive specific expression in these cells.
Collapse
MESH Headings
- 3T3 Cells
- Amino Acid Motifs
- Animals
- Base Sequence
- Cardiovascular System/embryology
- Cardiovascular System/growth & development
- Cardiovascular System/metabolism
- Cells, Cultured
- Consensus Sequence
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/pharmacology
- Desmin/genetics
- Fetal Heart/metabolism
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Genes
- Genes, Reporter
- Lac Operon
- Mice
- Mice, Transgenic
- Microfilament Proteins/deficiency
- Microfilament Proteins/genetics
- Microfilament Proteins/physiology
- Molecular Sequence Data
- Muscle Development
- Muscle Proteins/deficiency
- Muscle Proteins/genetics
- Muscle Proteins/physiology
- Muscle, Skeletal/embryology
- Muscle, Skeletal/growth & development
- Muscle, Skeletal/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Mutagenesis, Site-Directed
- Nuclear Proteins/metabolism
- Nuclear Proteins/pharmacology
- Promoter Regions, Genetic
- Regulatory Sequences, Nucleic Acid
- Serum Response Factor
- Transcription Factors/metabolism
- Transcription Factors/pharmacology
- Transcription, Genetic/drug effects
- Transcription, Genetic/genetics
- Transfection
- beta-Galactosidase/analysis
- beta-Galactosidase/genetics
Collapse
Affiliation(s)
- M Mericskay
- Laboratoire de Biologie Moléculaire de la Différenciation, Université Denis Diderot Paris 7, 2, place Jussieu, Paris, 75005, France
| | | | | | | | | | | | | |
Collapse
|
18
|
Kaji T, Yamada A, Miyajima S, Yamamoto C, Fujiwara Y, Wight TN, Kinsella MG. Cell density-dependent regulation of proteoglycan synthesis by transforming growth factor-beta(1) in cultured bovine aortic endothelial cells. J Biol Chem 2000; 275:1463-70. [PMID: 10625699 DOI: 10.1074/jbc.275.2.1463] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulation of vascular endothelial cell behavior during angiogenesis and in disease by transforming growth factor-beta(1) (TGF-beta(1)) is complex, but it clearly involves growth factor-induced changes in extracellular matrix synthesis. Proteoglycans (PGs) synthesized by endothelial cells contribute to the formation of the vascular extracellular matrix and also influence cellular proliferation and migration. Since the effects of TGF-beta(1) on vascular smooth muscle cell growth are dependent on cell density, it is possible that TGF-beta(1) also directs different patterns of PG synthesis in endothelial cells at different cell densities. In the present study, dense and sparse cultures of bovine aortic endothelial cells were metabolically labeled with [(3)H]glucosamine, [(35)S]sulfate, or (35)S-labeled amino acids in the presence of TGF-beta(1). The labeled PGs were characterized by DEAE-Sephacel ion exchange chromatography and Sepharose CL-4B molecular sieve chromatography. The glycosaminoglycan M(r) and composition were analyzed by Sepharose CL-6B chromatography, and the core protein M(r) was analyzed by SDS-polyacrylamide gel electrophoresis, before and after digestion with papain, heparitinase, or chondroitin ABC lyase. These experiments indicate that the effect of TGF-beta(1) on vascular endothelial cell PG synthesis is dependent on cell density. Specifically, TGF-beta(1) induced an accumulation of small chondroitin/dermatan sulfate PGs (CS/DSPGs) with core proteins of approximately 50 kDa in the medium of both dense and sparse cultures, but a cell layer-associated heparan sulfate PG with a core protein size of approximately 400 kDa accumulated only in dense cultures. Moreover, only in the dense cell cultures did TGF-beta(1) cause CS/DSPG hydrodynamic size to increase, which was due to the synthesis of CS/DSPGs with longer glycosaminoglycan chains. The heparan sulfate PG and CS/DSPG core proteins were identified as perlecan and biglycan, respectively, by Western blot analysis. The present data suggest that TGF-beta(1) promotes the synthesis of both perlecan and biglycan when endothelial cell density is high, whereas only biglycan synthesis is stimulated when the cell density is low. Furthermore, glycosaminoglycan chains are elongated only in biglycan synthesized by the cells at a high cell density.
Collapse
Affiliation(s)
- T Kaji
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Hokuriku University, Ho-3 Kanagawa-machi, Kanazawa 920-1181, Japan.
| | | | | | | | | | | | | |
Collapse
|
19
|
Weiser-Evans MC, Quinn BE, Burkard MR, Stenmark KR. Transient reexpression of an embryonic autonomous growth phenotype by adult carotid artery smooth muscle cells after vascular injury. J Cell Physiol 2000; 182:12-23. [PMID: 10567912 DOI: 10.1002/(sici)1097-4652(200001)182:1<12::aid-jcp2>3.0.co;2-g] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
High rates of vascular smooth muscle cell (SMC) replication are observed, at least transiently, after injury to the arterial wall and contribute to the formation of a neointima. Neutralizing antibodies designed to inhibit growth of SMC have only been variably successful in inhibiting neointima formation, raising the possibility that neointimal cell proliferation involves unique growth mechanisms. This study examined the possibility that SMC isolated from injured rat carotid arteries would express an autonomous, mitogen-independent growth phenotype similar to that utilized by embryonic vascular SMC during periods of rapid growth. We found that primary cultures of SMC isolated 7 and 14 days after injury, times at which high in vivo replication rates were observed, demonstrated high intrinsic DNA synthetic rates compared to SMC isolated from uninjured arteries or at 2, 4, 21, and 28 days after injury where in vivo replication rates were far less. Subcultured SMC isolated from 7-day injured vessels (Neo7 SMC) exhibited a stable, autonomous growth phenotype, did not secrete detectable mitogenic activity, and had decreased alpha-actin and myosin expression compared to mitogen-dependent SMC. Heterokaryons constructed between autonomous Neo7 SMC and mitogen-dependent SMC exhibited a mitogen-dependent growth phenotype suggesting that nonautonomous SMC produce factors that actively inhibit autonomous growth. In contrast, heterokaryons constructed between Neo7 SMC and autonomous embryonic SMC retained an autonomous growth phenotype. We examined the expression of known tumor suppressors to determine if any of these factors played a role in inhibiting SMC autonomous growth. p27, p53, pRb, and PTEN were abundantly expressed by Neo7 SMC and e17 SMC under both basal and serum stimulated conditions. The data suggest that the mechanisms driving SMC replication during neointimal formation are self-driven and self-regulated, and that at specific times after injury, SMC escape normal growth suppressive mechanisms through the loss of intracellular growth suppressor activity.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Aorta/drug effects
- Aorta/embryology
- Aorta/growth & development
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Catheterization
- Cell Division/drug effects
- Cell Fusion
- Cell Size/drug effects
- Cells, Cultured
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/pharmacology
- Female
- Genes, Tumor Suppressor/physiology
- Growth Substances/analysis
- Growth Substances/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Phenotype
- Proliferating Cell Nuclear Antigen/metabolism
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Rats
- Rats, Sprague-Dawley
- Tunica Intima/cytology
- Tunica Intima/drug effects
- Tunica Intima/injuries
- Tunica Intima/metabolism
Collapse
Affiliation(s)
- M C Weiser-Evans
- Department of Pediatrics, Developmental Lung Biology Laboratory, University of Colorado Health Sciences Center, Denver, Colorado, USA.
| | | | | | | |
Collapse
|
20
|
Paka L, Goldberg IJ, Obunike JC, Choi SY, Saxena U, Goldberg ID, Pillarisetti S. Perlecan mediates the antiproliferative effect of apolipoprotein E on smooth muscle cells. An underlying mechanism for the modulation of smooth muscle cell growth? J Biol Chem 1999; 274:36403-8. [PMID: 10593935 DOI: 10.1074/jbc.274.51.36403] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Apolipoprotein E (apoE) is known to inhibit cell proliferation; however, the mechanism of this inhibition is not clear. We recently showed that apoE stimulates endothelial production of heparan sulfate (HS) enriched in heparin-like sequences. Because heparin and HS are potent inhibitors of smooth muscle cell (SMC) proliferation, in this study we determined apoE effects on SMC HS production and cell growth. In confluent SMCs, apoE (10 microg/ml) increased (35)SO(4) incorporation into PG in media by 25-30%. The increase in the medium was exclusively due to an increase in HSPGs (2.2-fold), and apoE did not alter chondroitin and dermatan sulfate proteoglycans. In proliferating SMCs, apoE inhibited [(3)H]thymidine incorporation into DNA by 50%; however, despite decreasing cell number, apoE increased the ratio of (35)SO(4) to [(3)H]thymidine from 2 to 3.6, suggesting increased HS per cell. Purified HSPGs from apoE-stimulated cells inhibited cell proliferation in the absence of apoE. ApoE did not inhibit proliferation of endothelial cells, which are resistant to heparin inhibition. Analysis of the conditioned medium from apoE-stimulated cells revealed that the HSPG increase was in perlecan and that apoE also stimulated perlecan mRNA expression by >2-fold. The ability of apoE isoforms to inhibit cell proliferation correlated with their ability to stimulate perlecan expression. An anti-perlecan antibody completely abrogated the antiproliferative effect of apoE. Thus, these data show that perlecan is a potent inhibitor of SMC proliferation and is required to mediate the antiproliferative effect of apoE. Because other growth modulators also regulate perlecan expression, this may be a key pathway in the regulation of SMC growth.
Collapse
Affiliation(s)
- L Paka
- Department of Radiation Oncology, North Shore-Long Island Jewish Health System, Manhasset, New York 11030, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Whitelock JM, Graham LD, Melrose J, Murdoch AD, Iozzo RV, Underwood PA. Human perlecan immunopurified from different endothelial cell sources has different adhesive properties for vascular cells. Matrix Biol 1999; 18:163-78. [PMID: 10372557 DOI: 10.1016/s0945-053x(99)00014-1] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Perlecan, a major heparan sulfate proteoglycan of vascularized tissues, was immunopurified from media conditioned by human endothelial cells of both arterial and venous origin. The heparan sulfate moiety of perlecan from cultured arterial cells differed in amount and/or composition from that produced by a transformed cell line of venous origin. Both forms of perlecan bound basic fibroblast growth factor with Kd approximately 70 nM. In ELISA experiments, perlecan and its protein core bound to various extracellular matrix components in a manner that was strongly influenced by the format of the assay. Human vascular smooth muscle cells and human endothelial cells adhered to perlecan-coated surfaces, and both cell types adhered better to the venous cell-derived than to the arterial cell-derived perlecan. Removal of the heparan sulfate chains abolished this difference and increased the ability of both types of perlecan to adhere vascular cells. Denaturation of perlecan and its protein core also rendered each of them more adhesive, indicating the presence of conformation-independent adhesion determinants in the polypeptide sequence. Their location was investigated using recombinant perlecan domains. Overall, our results represent the first demonstration of human perlecan acting as an adhesive molecule for human vascular cells and suggest that it may play a role in vascular wound healing.
Collapse
Affiliation(s)
- J M Whitelock
- Co-operative Research Center for Cardiac Technology, North Ryde, NSW, Australia.
| | | | | | | | | | | |
Collapse
|
22
|
Iozzo RV, Danielson KG. Transcriptional and posttranscriptional regulation of proteoglycan gene expression. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 1999; 62:19-53. [PMID: 9932451 DOI: 10.1016/s0079-6603(08)60504-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Proteoglycans are among the most complex and sophisticated molecules of mammalian systems in terms of their protein and carbohydrate moieties. These macromolecules are in a continuous interplay with each other and the cell surface signal-transducing pathways, some of which are beginning to be elucidated. Because of their domain structure, catalytic potential, and diversity, these molecules appear to be designed for integrating numerous signaling events. For example, some proteoglycans interact with hyaluronan and lectins, thereby linking cell surfaces and distant matrix molecules. Some interact with collagen during the complex process of fibrillogenesis and regulate this biological process fundamental to animal life. Others interact with growth factors and serve as depot available during growth or tissue remodeling. In this review, we center on the most recent developments of proteoglycan biology, focusing primarily on genomic organization and transcriptional and posttranscriptional control. We discuss only those proteoglycans whose gene and promoter elements have been characterized and proved to be functional. When possible, we correlate the effects of growth factors and cytokines on proteoglycan gene expression with the topology of cis-acting elements in their genomic control regions. The analysis leads to a comprehensive critical appraisal of the principles that underlie the regulation of proteoglycan gene expression and to the delineation of common regulatory mechanisms.
Collapse
Affiliation(s)
- R V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | |
Collapse
|
23
|
Belknap JK, Weiser-Evans MC, Grieshaber SS, Majack RA, Stenmark KR. Relationship between perlecan and tropoelastin gene expression and cell replication in the developing rat pulmonary vasculature. Am J Respir Cell Mol Biol 1999; 20:24-34. [PMID: 9870914 DOI: 10.1165/ajrcmb.20.1.3321] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Smooth-muscle-cell (SMC) replication and extracellular matrix protein expression are two vital and interrelated processes necessary for normal development of the vasculature. To understand better the nature of this relationship in the developing rat lung, we investigated the relationship between SMC proliferation and the expression of perlecan, a basement membrane (BM) heparan sulfate proteoglycan implicated in the control of SMC growth and differentiation, and tropoelastin (TE), a structural matrix protein not known to influence directly the replicative state of SMCs. Using bromodeoxyuridine (BrdU) incorporation to assess DNA synthesis, we first established the time course of SMC proliferation in the hilar pulmonary artery (PA) from embryonic to adult life. We found a labeling index of > 80% during the embryonic period (embryonic Day 13 [e13] to fetal Day 18 [f18]), a dramatic decline to approximately 40% during the fetal period of development, and a steady decrease in proliferation rates following birth such that, by 30 d of age, a labeling index of < 2% was noted. Using in situ hybridization, we found that although peak expression of both perlecan and TE messenger RNA (mRNA) occurred in the fetal and early postnatal periods following the major decrease in cell replication, TE mRNA expression was clearly observed in the PA as early as embryonic Day 14, whereas perlecan transcripts were virtually undetectable until fetal Day 19. Therefore, to evaluate further the relationship between cell replication and perlecan and/or TE gene expression, we used a combined in situ hybridization/BrdU immunohistochemistry technique and demonstrated that, on an individual cell basis, perlecan message was predominantly expressed by nonreplicating (BrdU-negative) PA, whereas TE mRNA was equally expressed in replicating and nonreplicating PA SMCs. Interestingly, a very similar pattern of replication and relationship to perlecan and TE mRNA expression was noted in airway SMCs and epithelial cells. Thus, in the lung as a whole, maximal expression of both the BM protein perlecan and the interstitial matrix protein TE occurs coordinately and follows the period of maximal SMC proliferation. However, in individual SMCs, perlecan mRNA expression varies inversely with DNA synthesis, whereas TE mRNA expression appears independent of the proliferative state of the cell.
Collapse
Affiliation(s)
- J K Belknap
- Department of Pediatrics, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | | | | | |
Collapse
|
24
|
Abstract
The proteoglycan superfamily now contains more than 30 full-time molecules that fulfill a variety of biological functions. Proteoglycans act as tissue organizers, influence cell growth and the maturation of specialized tissues, play a role as biological filters and modulate growth-factor activities, regulate collagen fibrillogenesis and skin tensile strength, affect tumor cell growth and invasion, and influence corneal transparency and neurite outgrowth. Additional roles, derived from studies of mutant animals, indicate that certain proteoglycans are essential to life whereas others might be redundant. The review focuses on the most recent genetic and molecular biological studies of the matrix proteoglycans, broadly defined as proteoglycans secreted into the pericellular matrix. Special emphasis is placed on the molecular organization of the protein core, the utilization of protein modules, the gene structure and transcriptional control, and the functional roles of the various proteoglycans. When possible, proteoglycans have been grouped into distinct gene families and subfamilies offering a simplified nomenclature based on their protein core design. The structure-function relationship of some paradigmatic proteoglycans is discussed in depth and novel aspects of their biology are examined.
Collapse
Affiliation(s)
- R V Iozzo
- Department of Pathology, Anatomy and Cell Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107-6799, USA.
| |
Collapse
|
25
|
Koyama N, Kinsella MG, Wight TN, Hedin U, Clowes AW. Heparan sulfate proteoglycans mediate a potent inhibitory signal for migration of vascular smooth muscle cells. Circ Res 1998; 83:305-13. [PMID: 9710123 DOI: 10.1161/01.res.83.3.305] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Migration of vascular smooth muscle cells (SMCs) is a key step in vascular remodeling and formation of pathological lesions in diseased arteries and may be controlled by extracellular matrix (ECM) and by factors that regulate ECM composition, such as platelet-derived growth factor (PDGF). In culture, PDGF-AB and -BB enhance but PDGF-AA (although having no effect alone) suppresses SMC migration stimulated by other PDGF isoforms. To determine whether the migration-inhibitory mechanism of PDGF-AA was mediated by ECM composition, we examined baboon SMC migration in a Boyden chamber assay using filters coated with different ECM proteins. PDGF-AA suppressed the PDGF-BB-induced migration of baboon SMCs on a filter coated with basement membrane proteins (Matrigel) and fibronectin but failed to inhibit cell migration on a type I collagen (Vitrogen)-coated filter. Fibronectin and fibronectin fragments that contain heparin-binding domains permitted PDGF-AA inhibition of cell migration, but a fragment lacking heparin-binding domains did not. Treatment of SMCs with heparin lyases II and III, but not with chondroitin ABC lyase, diminished the PDGF-AA-mediated inhibition of migration. PDGF-AA stimulated accumulation of proteoglycan (PG) in the cell layer more potently than did PDGF-BB, whereas the turnover of cell layer PG was unaffected by either PDGF-AA or -BB. Northern blot analysis revealed that PDGF-AA increased syndecan-1 mRNA expression more than did PDGF-BB, whereas both PDGF isoforms decreased perlecan expression. The changes in cell migration and PG synthesis induced by PDGF-AA were accompanied by changes in the morphology of SMCs. PDGF-AA dramatically induced the spreading of SMCs, whereas the heparin lyase treatment of PDGF-AA-stimulated cultures diminished cell spreading. The data suggest that PDGF-AA selectively modifies heparan sulfate PG accumulation on SMCs and thereby influences the interactions of SMCs with heparin-binding ECM proteins. These interactions, in turn, generate signals that suppress SMC migration.
Collapse
Affiliation(s)
- N Koyama
- Department of Surgery, University of Washington, Seattle 98195-6410, USA
| | | | | | | | | |
Collapse
|
26
|
Evanko SP, Raines EW, Ross R, Gold LI, Wight TN. Proteoglycan distribution in lesions of atherosclerosis depends on lesion severity, structural characteristics, and the proximity of platelet-derived growth factor and transforming growth factor-beta. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 152:533-46. [PMID: 9466580 PMCID: PMC1857967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The accumulation of proteoglycans (PGs) in atherosclerosis contributes to disease progression and stenosis and may partly depend on local regulation by growth factors such as platelet-derived growth factor (PDGF) and transforming growth factor (TGF)-beta. In this study, the distribution of the major extracellular PGs is compared with that of PDGF and TGF-beta isoforms in developing lesions of atherosclerosis from hypercholesterolemic nonhuman primates. Strong immunostaining for decorin, biglycan, versican, and hyaluronan is observed in both intermediate and advanced lesions. Perlecan staining is weak in intermediate lesions but strong in advanced lesions in areas bordering the plaque core. Immunostaining for PDGF-B and TGF-beta1 is particularly prominent in macrophages in intermediate and advanced lesions. In contrast, TGF-beta2 and TGF-beta3 and PDGF-A are present in both macrophages and smooth muscle cells. Overall, PG deposits parallel areas of intense growth factor immunostaining, with trends in relative localization that suggest interrelationships among certain PGs and growth factors. Notably, decorin and TGF-beta1 are distributed similarly, predominantly in the macrophage-rich core, whereas biglycan is prominent in the smooth muscle cell matrix adjoining TGF-beta1-positive macrophages. Versican and hyaluronan are enriched in the extracellular matrix adjacent to both PDGF- and TGF-beta1-positive cells. These data demonstrate that PG accumulation varies with lesion severity, structural characteristics, and the proximity of PDGF and TGF-beta.
Collapse
Affiliation(s)
- S P Evanko
- Department of Pathology, University of Washington, Seattle 98195, USA
| | | | | | | | | |
Collapse
|