1
|
Ballantyne MD, McDonald RA, Baker AH. lncRNA/MicroRNA interactions in the vasculature. Clin Pharmacol Ther 2016; 99:494-501. [PMID: 26910520 PMCID: PMC4881297 DOI: 10.1002/cpt.355] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/16/2016] [Indexed: 12/14/2022]
Abstract
MicroRNA (miRNA) have gained widespread attention for their role in diverse vascular processes including angiogenesis, apoptosis, proliferation, and migration. Despite great understanding of miRNA expression and function, knowledge of long noncoding RNA (lncRNA) molecular mechanisms still remains limited. The influence of miRNA on lncRNA function, and the converse, is now beginning to emerge. lncRNA may regulate miRNA function by acting as endogenous sponges to regulate gene expression and miRNA have been shown to bind and regulate lncRNA stability. A detailed understanding of the molecular and cellular effects of lncRNA‐miRNA‐mediated interactions in vascular pathophysiology could pave the way for new diagnostic markers and therapeutic approaches, but first there is a requirement for a more detailed understanding of the impact of such regulatory networks.
Collapse
Affiliation(s)
- M D Ballantyne
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK.,Institute of Cardiovascular and Medical Science, British Heart Foundation Glasgow, Glasgow, UK
| | - R A McDonald
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - A H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
2
|
Unseld M, Chilla A, Pausz C, Mawas R, Breuss J, Zielinski C, Schabbauer G, Prager GW. PTEN expression in endothelial cells is down-regulated by uPAR to promote angiogenesis. Thromb Haemost 2015; 114:379-89. [PMID: 25925849 DOI: 10.1160/th15-01-0016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/16/2015] [Indexed: 12/16/2022]
Abstract
The tumour suppressor phosphatase and tensin homologue (PTEN), mutated or lost in many human cancers, is a major regulator of angiogenesis. However, the cellular mechanism of PTEN regulation in endothelial cells so far remains elusive. Here, we characterise the urokinase receptor (uPAR, CD87) and its tumour-derived soluble form, suPAR, as a key molecule of regulating PTEN in endothelial cells. We observed uPAR-deficient endothelial cells to express enhanced PTEN mRNA- and protein levels. Consistently, uPAR expression in endogenous negative uPAR cells, down-regulated PTEN and activated the PI3K/Akt pathway. Additionally, we found that integrin adhesion receptors act as trans-membrane signaling partners for uPAR to repress PTEN transcription in a NF-κB-dependent manner. Functional in vitro assays with endothelial cells, derived from uPAR-deficient and PTEN heterozygous crossbred mice, demonstrated the impact of uPAR-dependent PTEN regulation on cell motility and survival. In an in vivo murine angiogenesis model uPAR-deficient PTEN heterozygous animals increased the impaired angiogenic phenotype of uPAR knockout mice and were able to reverse the high invasive potential of PTEN heterozygots. Our data provide first evidence that endogenous as well as exogenous soluble uPAR down-regulated PTEN in endothelial cells to support angiogenesis. The uPAR-induced PTEN regulation might represent a novel target for drug interference, and may lead to the development of new therapeutic strategies in anti-angiogenic treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - G Schabbauer
- Gernot Schabbauer, Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria, Tel.: +43 1 40160 31427, Fax: +43 1 40160 93101, E-mail:
| | - G W Prager
- Gerald W. Prager, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria, Tel.: +43 1 40400 4450, Fax: +43 1 40400 4451, e-mail:
| |
Collapse
|
3
|
Gilder AS, Jones KA, Hu J, Wang L, Chen CC, Carter BS, Gonias SL. Soluble Urokinase Receptor Is Released Selectively by Glioblastoma Cells That Express Epidermal Growth Factor Receptor Variant III and Promotes Tumor Cell Migration and Invasion. J Biol Chem 2015; 290:14798-809. [PMID: 25837250 DOI: 10.1074/jbc.m115.637488] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 12/13/2022] Open
Abstract
Genomic heterogeneity is characteristic of glioblastoma (GBM). In many GBMs, the EGF receptor gene (EGFR) is amplified and may be truncated to generate a constitutively active form of the receptor called EGFRvIII. EGFR gene amplification and EGFRvIII are associated with GBM progression, even when only a small fraction of the tumor cells express EGFRvIII. In this study, we show that EGFRvIII-positive GBM cells express significantly increased levels of cellular urokinase receptor (uPAR) and release increased amounts of soluble uPAR (suPAR). When mice were xenografted with human EGFRvIII-expressing GBM cells, tumor-derived suPAR was detected in the plasma, and the level was significantly increased compared with that detected in plasma samples from control mice xenografted with EGFRvIII-negative GBM cells. suPAR also was increased in plasma from patients with EGFRvIII-positive GBMs. Purified suPAR was biologically active when added to cultures of EGFRvIII-negative GBM cells, activating cell signaling and promoting cell migration and invasion. suPAR did not significantly stimulate cell signaling or migration of EGFRvIII-positive cells, probably because cell signaling was already substantially activated in these cells. The activities of suPAR were replicated by conditioned medium (CM) from EGFRvIII-positive GBM cells. When the CM was preincubated with uPAR-neutralizing antibody or when uPAR gene expression was silenced in cells used to prepare CM, the activity of the CM was significantly attenuated. These results suggest that suPAR may function as an important paracrine signaling factor in EGFRvIII-positive GBMs, inducing an aggressive phenotype in tumor cells that are EGFRvIII-negative.
Collapse
Affiliation(s)
| | | | | | - Lei Wang
- From the Departments of Pathology and
| | - Clark C Chen
- Surgery, Division of Neurosurgery, University of California at San Diego, La Jolla, California 92093
| | - Bob S Carter
- Surgery, Division of Neurosurgery, University of California at San Diego, La Jolla, California 92093
| | | |
Collapse
|
4
|
Laçin NT, Utkan GG. Role of biomaterials in prevention of in-stent restenosis. J Biomed Mater Res B Appl Biomater 2013; 102:1113-20. [PMID: 24307479 DOI: 10.1002/jbm.b.33083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 05/30/2013] [Accepted: 11/19/2013] [Indexed: 01/01/2023]
Abstract
Coronary balloon angioplasty and coronary stenting are the procedures used in healing coronary artery disease. However, injury of arteries during angioplasty and stenting causes cell stimulations in tissue. Cell movement and thrombosis lead to re-narrowing of widened vessel called restenosis. Several new types of carriers and technology have been developed to suppress and/or prevent restenosis. Authors review the polymeric materials featured in drug/gene carrier systems, nanovehicles, and stent coating materials against restenosis.
Collapse
Affiliation(s)
- Nelisa T Laçin
- Advanced Technology Education, Research and Application Center, Mersin University, 33343, Mersin, Turkey
| | | |
Collapse
|
5
|
Garrido-Martín EM, Blanco FJ, Roquè M, Novensà L, Tarocchi M, Lang UE, Suzuki T, Friedman SL, Botella LM, Bernabéu C. Vascular injury triggers Krüppel-like factor 6 mobilization and cooperation with specificity protein 1 to promote endothelial activation through upregulation of the activin receptor-like kinase 1 gene. Circ Res 2012; 112:113-27. [PMID: 23048070 DOI: 10.1161/circresaha.112.275586] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RATIONALE Activin receptor-like kinase-1 (ALK1) is an endothelial transforming growth factor β receptor involved in angiogenesis. ALK1 expression is high in the embryo vasculature, becoming less detectable in the quiescent endothelium of adult stages. However, ALK1 expression becomes rapidly increased after angiogenic stimuli such as vascular injury. OBJECTIVE To characterize the molecular mechanisms underlying the regulation of ALK1 on vascular injury. METHODS AND RESULTS Alk1 becomes strongly upregulated in endothelial (EC) and vascular smooth muscle cells of mouse femoral arteries after wire-induced endothelial denudation. In vitro denudation of monolayers of human umbilical vein ECs also leads to an increase in ALK1. Interestingly, a key factor in tissue remodeling, Krüppel-like factor 6 (KLF6) translocates to the cell nucleus during wound healing, concomitantly with an increase in the ALK1 gene transcriptional rate. KLF6 knock down in human umbilical vein ECs promotes ALK1 mRNA downregulation. Moreover, Klf6(+/-) mice have lower levels of Alk1 in their vasculature compared with their wild-type siblings. Chromatin immunoprecipitation assays show that KLF6 interacts with ALK1 promoter in ECs, and this interaction is enhanced during wound healing. We demonstrate that KLF6 is transactivating ALK1 gene, and this transactivation occurs by a synergistic cooperative mechanism with specificity protein 1. Finally, Alk1 levels in vascular smooth muscle cells are not directly upregulated in response to damage, but in response to soluble factors, such as interleukin 6, released from ECs after injury. CONCLUSIONS ALK1 is upregulated in ECs during vascular injury by a synergistic cooperative mechanism between KLF6 and specificity protein 1, and in vascular smooth muscle cells by an EC-vascular smooth muscle cell paracrine communication during vascular remodeling.
Collapse
|
6
|
Benza RL, Passineau MJ, Anderson PG, Barchue JP, George JF. The role of fibrinolytic genes and proteins in the development of allograft vascular disease. J Heart Lung Transplant 2011; 30:935-44. [PMID: 21652221 DOI: 10.1016/j.healun.2011.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 04/19/2011] [Accepted: 04/19/2011] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND We have previously shown that lack of plasminogen activator inhibitor-1 (PAI-1) expression in donor tissue greatly increases intimal proliferation (IP) after allogeneic transplantation. We sought to determine the relative role of PAI-1 and other fibrinolytic proteins in the development of IP. METHODS We used an abdominal aortic transplant model in mice to investigate IP in 3 groups of 6 recipients. In the isograft group, CBA/J strain mice were donors and recipients, donors for allograft group were C57BL/6J mice, and for the allograft/knockout group, C57BL/6J PAI-1 knockout mice. All groups received weekly injections of anti-CD8/CD4 monoclonal antibodies. IP was calculated at 50 days, and sections were analyzed for fibrinolytic proteins, messenger RNA (mRNA) and PAI-1 activity using immunohistochemistry (IHC), in situ hybridization (ISH), reverse transcription-polymerase chain reaction (RT-PCR), and Western blot analysis. RESULTS Significantly more IP developed in the allograft/knockout group vs the isograft (p < 0.001) and the allograft groups (p = 0.003). There was marked intimal expression of tissue plasminogen activator (tPA), urokinase PA (uPA), and uPA receptor (uPAR) proteins and mRNA in the allograft and allograft/knockout groups vs the isograft group. Allografts also showed significant intimal staining for PAI-1 protein and mRNA. RT-PCR demonstrated a stepwise increase in profibrinolytic protein mRNA from isograft to allograft to allograft/knockout groups, particularly uPA (p = 0.02) and uPAR (p = 0.016). Western blot data showed complementary findings. PAI-1 activity was persistently present in isograft and allograft animals, only. Intimas in allograft and allograft/knockout groups were primarily smooth muscle cells. CONCLUSIONS PAI-1 reduces IP by limiting smooth muscle cell activity, with little change in matrix composition likely by modulating profibrinolytic protein expression.
Collapse
Affiliation(s)
- Raymond L Benza
- Division of Cardiovascular Medicine, Allegheny General Hospital, Pittsburgh, Pennsylvania 15212, USA.
| | | | | | | | | |
Collapse
|
7
|
Dusio GF, Cardani D, Zanobbio L, Mantovani M, Luchini P, Battini L, Galli V, Diana A, Balsari A, Rumio C. Stimulation of TLRs by LMW-HA induces self-defense mechanisms in vaginal epithelium. Immunol Cell Biol 2010; 89:630-9. [PMID: 21102537 DOI: 10.1038/icb.2010.140] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The innate immune system is present throughout the female reproductive tract and functions in synchrony with the adaptive immune system to provide protection in a way that enhances the chances for fetal survival, while protecting against potential pathogens. Recent data show that activation of Toll-like receptor (TLR)2 and 4 by low-molecular weight hyaluronic acid (LMW-HA) in the epidermis induces secretion of the antimicrobial peptide β-defensin 2. In the present work, we show that LMW-HA induces vaginal epithelial cells to release different antimicrobial peptides, via activation of TLR2 and TLR4. Further, we found that LMW-HA favors repair of vaginal epithelial injury, involving TLR2 and TLR4, and independently from its classical receptor CD44. This wound-healing activity of LMW-HA is dependent from an Akt/phosphatidylinositol 3 kinase pathway. Therefore, these findings suggest that the vaginal epithelium is more than a simple physical barrier to protect against invading pathogens: on the contrary, this surface acts as efficient player of innate host defense, which may modulate its antimicrobial properties and injury restitution activity, following LMW-HA stimulation; this activity may furnish an additional protective activity to this body compartment, highly and constantly exposed to microbiota, ameliorating the self-defense of the vaginal epithelium in both basal and pathological conditions.
Collapse
Affiliation(s)
- Giuseppina F Dusio
- iMIL, italian Mucosal Immunity Laboratory, Dipartimento di Morfologia Umana e Scienze Biomediche Città Studi, Università degli Studi di Milano, Milano, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Orlandi A, Ferlosio A, Arcuri G, Scioli MG, De Falco S, Spagnoli LG. Flt-1 expression influences apoptotic susceptibility of vascular smooth muscle cells through the NF-κB/IAP-1 pathway. Cardiovasc Res 2009; 85:214-23. [DOI: 10.1093/cvr/cvp288] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
9
|
Tkachuk VA, Plekhanova OS, Parfyonova YV. Regulation of arterial remodeling and angiogenesis by urokinase-type plasminogen activatorThis article is one of a selection of papers from the NATO Advanced Research Workshop on Translational Knowledge for Heart Health (published in part 2 of a 2-part Special Issue). Can J Physiol Pharmacol 2009; 87:231-51. [DOI: 10.1139/y08-113] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A wide variety of disorders are associated with an imbalance in the plasminogen activator system, including inflammatory diseases, atherosclerosis, intimal hyperplasia, the response mechanism to vascular injury, and restenosis. Urokinase-type plasminogen activator (uPA) is a multifunctional protein that in addition to its fibrinolytic and matrix degradation capabilities also affects growth factor bioavailability, cytokine modulation, receptor shedding, cell migration and proliferation, phenotypic modulation, protein expression, and cascade activation of proteases, inhibitors, receptors, and modulators. uPA is the crucial protein for neointimal growth and vascular remodeling. Moreover, it was recently shown to be implicated in the stimulation of angiogenesis, which makes it a promising multipurpose therapeutic target. This review is focused on the mechanisms by which uPA can regulate arterial remodeling, angiogenesis, and cell migration and proliferation after arterial injury and the means by which it modulates gene expression in vascular cells. The role of domain specificity of urokinase in these processes is also discussed.
Collapse
Affiliation(s)
- Vsevolod A. Tkachuk
- Cardiology Research Centre, Laboratory of Molecular Endocrinology, Moscow 121552, Russia
- Medical School, Lomonosov Moscow State University, Moscow, Russia
| | - Olga S. Plekhanova
- Cardiology Research Centre, Laboratory of Molecular Endocrinology, Moscow 121552, Russia
- Medical School, Lomonosov Moscow State University, Moscow, Russia
| | - Yelena V. Parfyonova
- Cardiology Research Centre, Laboratory of Molecular Endocrinology, Moscow 121552, Russia
- Medical School, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
10
|
Anea CB, Zhang M, Stepp DW, Bryan Simkins G, Reed G, Fulton DJ, Daniel Rudic R. Vascular disease in mice with a dysfunctional circadian clock. Circulation 2009; 119:1510-7. [PMID: 19273720 PMCID: PMC2761686 DOI: 10.1161/circulationaha.108.827477] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiovascular disease is the leading cause of death for both men and women in the United States and the world. A profound pattern exists in the time of day at which the death occurs; it is in the morning, when the endothelium is most vulnerable and blood pressure surges, that stroke and heart attack most frequently happen. Although the molecular components of circadian rhythms rhythmically oscillate in blood vessels, evidence of a direct function for the "circadian clock" in the progression to vascular disease is lacking. METHODS AND RESULTS In the present study, we found increased pathological remodeling and vascular injury in mice with aberrant circadian rhythms, Bmal1-knockout and Clock mutant. In addition, naive aortas from Bmal1-knockout and Clock mutant mice exhibit endothelial dysfunction. Akt and subsequent nitric oxide signaling, a pathway critical to vascular function, was significantly attenuated in arteries from Bmal1-knockout mice. CONCLUSIONS Our data reveal a new role for the circadian clock during chronic vascular responses that may be of significance in the progression of vascular disease.
Collapse
Affiliation(s)
- Ciprian B. Anea
- Department of Pharmacology & Toxicology (C.B.A., M.Z., G.B.S., D.J.F., R.D.R.); Department of Physiology (D.W.S.); Vascular Biology Center (D.W.S., D.J.F.); and the Cardiology Division, Department of Medicine (G.R.), Medical College of Georgia, Augusta, GA
| | - Maoxiang Zhang
- Department of Pharmacology & Toxicology (C.B.A., M.Z., G.B.S., D.J.F., R.D.R.); Department of Physiology (D.W.S.); Vascular Biology Center (D.W.S., D.J.F.); and the Cardiology Division, Department of Medicine (G.R.), Medical College of Georgia, Augusta, GA
| | - David W. Stepp
- Department of Pharmacology & Toxicology (C.B.A., M.Z., G.B.S., D.J.F., R.D.R.); Department of Physiology (D.W.S.); Vascular Biology Center (D.W.S., D.J.F.); and the Cardiology Division, Department of Medicine (G.R.), Medical College of Georgia, Augusta, GA
| | - G. Bryan Simkins
- Department of Pharmacology & Toxicology (C.B.A., M.Z., G.B.S., D.J.F., R.D.R.); Department of Physiology (D.W.S.); Vascular Biology Center (D.W.S., D.J.F.); and the Cardiology Division, Department of Medicine (G.R.), Medical College of Georgia, Augusta, GA
| | - Guy Reed
- Department of Pharmacology & Toxicology (C.B.A., M.Z., G.B.S., D.J.F., R.D.R.); Department of Physiology (D.W.S.); Vascular Biology Center (D.W.S., D.J.F.); and the Cardiology Division, Department of Medicine (G.R.), Medical College of Georgia, Augusta, GA
| | - David J. Fulton
- Department of Pharmacology & Toxicology (C.B.A., M.Z., G.B.S., D.J.F., R.D.R.); Department of Physiology (D.W.S.); Vascular Biology Center (D.W.S., D.J.F.); and the Cardiology Division, Department of Medicine (G.R.), Medical College of Georgia, Augusta, GA
| | - R. Daniel Rudic
- Department of Pharmacology & Toxicology (C.B.A., M.Z., G.B.S., D.J.F., R.D.R.); Department of Physiology (D.W.S.); Vascular Biology Center (D.W.S., D.J.F.); and the Cardiology Division, Department of Medicine (G.R.), Medical College of Georgia, Augusta, GA
| |
Collapse
|
11
|
Induction of Indefinite Cardiac Allograft Survival Correlates With Toll-Like Receptor 2 and 4 Downregulation After Serine Protease Inhibitor-1 (Serp-1) Treatment. Transplantation 2007; 84:1158-67. [DOI: 10.1097/01.tp.0000286099.50532.b0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Pawlak K, Pawlak D, Myśliwiec M. Urokinase-type plasminogen activator and metalloproteinase-2 are independently related to the carotid atherosclerosis in haemodialysis patients. Thromb Res 2007; 121:543-8. [PMID: 17706748 DOI: 10.1016/j.thromres.2007.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2007] [Revised: 06/04/2007] [Accepted: 06/19/2007] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Matrix metalloproteinases (MMPs), their tissue inhibitors (TIMPs) system, and fibrinolytic system, have been implicated as important factors in atherosclerosis and vascular remodeling. However, no data are yet available on the associations between these two systems in relation to carotid atherosclerosis in hemodialysis (HD) patients. MATERIAL AND METHODS We compared plasma levels of MMP-2, MMP-9, TIMP-1, TIMP-2; the parameters of fibrinolytic system: tissue-type plasminogen activator (tPA), urokinase-type plasminogen activator (uPAR) and its soluble receptor (suPAR), plasminogen activator inhibitor-1 (PAI-1), plasmin-alpha2-antiplasmin (PAP) complexes; high sensitivity C-reactive protein (hs CRP) as a marker of inflammation and a surrogate of atherosclerotic disease-intima media thickness (IMT) in HD patients and in healthy controls. RESULTS The values of the uPA, suPAR, PAP, MMP-2, TIMPs, hs CRP and IMT in the patients significantly exceeded those in controls. The concentrations of MMP-9, tPA and PAI-1 were similar in both investigated groups. uPA, uPAR and PAP were positively associated with MMP-2/TIMPs system; all mentioned above parameters (except TIMP-2) and hsCRP were associated with IMT. Multivariate analysis showed that uPA, MMP-2 and age were the strong independent variables linked to IMT values in HD patients. CONCLUSIONS The patients on haemodialysis treatment have evidence of disordered fibrinolysis/proteolysis balance in the plasma, independently associated with IMT on multivariate analysis. These data suggest the importance of uPA and MMP-2 levels in the developing of atherosclerosis in these patients.
Collapse
Affiliation(s)
- Krystyna Pawlak
- Department of Nephrology and Clinical Transplantation, Medical University, Bialystok, Poland.
| | | | | |
Collapse
|
13
|
Galaria II, Nicholl SM, Roztocil E, Davies MG. Urokinase-induced smooth muscle cell migration requires PI3-K and Akt activation. J Surg Res 2005; 127:46-52. [PMID: 15964303 DOI: 10.1016/j.jss.2005.02.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2004] [Revised: 02/22/2005] [Accepted: 02/23/2005] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To examine the role of the phospho-inositol-3'-kinase (PI3-K)-akt signaling axis during smooth muscle cell (SMC) migration in response to the aminoterminal fragment of urokinase (ATF). BACKGROUND Urokinase (uPA) is involved in vessel remodeling and mediates smooth muscle cell migration. Migration in response to urokinase is dependent on ATF. The role of PI3-K/akt signaling during migration in response to the uPA fragments is not understood. METHODS Murine arterial SMCs were cultured in vitro. Linear wound and Boyden microchemotaxis assays of migration were performed in the presence of ATF with and without the PI3-K inhibitors (Wortmannin, Wn [10 nm] and LY294002, LY [10 microm]) and an akt inhibitor (aktI, [10 microm]). Western blotting was performed for akt, ERK1/2, and GSK3beta phosphorylation after cells were stimulated with ATF in the presence and absence of the inhibitors. Statistics were analyzed by one-way ANOVA. RESULTS Both PI3-K and akt inhibitors blocked the migratory response to ATF in both assays. ATF induced time-dependent increases in akt phosphorylation at both S472 and T308 sites and ERK1/2 phosphorylation. Activation of akt and ERK1/2 was inhibited by Wn and LY. Manumycin A, a ras inhibitor, did not inhibit activation of akt but did inhibit ERK1/2 activation. Activation of akt and the dephosphorylation of its downstream kinase GSK3beta were inhibited by the akt inhibitor. Direct inhibition of akt did not influence ERK1/2 activation and inhibition of ERK1/2 did not influence akt activation. CONCLUSION ATF mediated migration is PI3-K dependent and activates two separate pathways: ERK1/2 and akt. ATF induces akt phosphorylation through a PI3K-mediated but ras-independent mechanism while both ras and PI3K are required for ERK1/2 activation. Defining key signaling pathways is vital to regulate vessel remodeling.
Collapse
Affiliation(s)
- Irfan I Galaria
- Division of Vascular Surgery, Department of Surgery, Center for Cardiovascular Research, University of Rochester, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|
14
|
Wang Z, Sosne G, Kurpakus-Wheater M. Plasminogen activator inhibitor-1 (PAI-1) stimulates human corneal epithelial cell adhesion and migration in vitro. Exp Eye Res 2005; 80:1-8. [PMID: 15652520 DOI: 10.1016/j.exer.2004.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2004] [Indexed: 10/26/2022]
Abstract
In addition to its role as an inhibitor of urokinase plasminogen activator (uPA), plasminogen activator inhibitor-1 (PAI-1) is hypothesized to regulate epithelial cell adhesion and migration. We have previously reported that PAI-1 may be an important regulatory factor of the uPA system in cornea. The purpose of this study was to extend those observations by determining the effect of exogenous PAI-1 on the migration and adhesion of human corneal epithelial cells (HCEC) in vitro. The expression of PAI-1 in non-transformed early passage HCEC was confirmed by immunofluorescence microscopy and Western blot analysis. Colorimetric assays coupled with function-inhibiting antibody studies using the matrix assembled in situ by cultured cells demonstrate that immobilized PAI-1 serves as an efficient substrate for HCEC adhesion. HCEC attachment to PAI-1 is comparable to that of laminin-10, a known strong adhesion protein for epithelial cells. In addition to serving as an adhesion substrate, PAI-1 also functions as a chemotactic agent for corneal epithelium. Additionally it promotes the random migration of HCEC, from an initial cell cluster, along a culture substrate. Our results in corneal epithelium are consistent with reports from other investigators showing that PAI-1 facilitates both epithelial adhesion and migration. From our studies we conclude that PAI-1 may play a dual role in corneal wound healing. Initially PAI-1 may function to stimulate migration and facilitate the reepithelialization of the wound bed. Post-reepithelization, PAI-1 may ensure corneal epithelial cell adhesion to matrix to promote successful wound healing.
Collapse
Affiliation(s)
- Zhiyu Wang
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | |
Collapse
|
15
|
Christ G, Nikfardjam M, Huber-Beckmann R, Gottsauner-Wolf M, Glogar D, Binder BR, Wojta J, Huber K. Predictive value of plasma plasminogen activator inhibitor-1 for coronary restenosis: dependence on stent implantation and antithrombotic medication. J Thromb Haemost 2005; 3:233-9. [PMID: 15670026 DOI: 10.1111/j.1538-7836.2004.01062.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The plasmin activation system is involved in the development of restenosis after percutaneous coronary interventions (PCI). Conflicting data exist concerning the role of plasminogen activator inhibitor-1 (PAI-1) and its predictive value for restenosis. OBJECTIVES To evaluate the fibrinolytic response to injury after PCI with or without stent implantation on different antithrombotic medications and its relation to late restenosis. PATIENTS AND METHODS Eighty consecutive patients with successful PCI without (balloon only; n = 37) or with stent implantation (stent; n = 43) on different antithrombotic regimes (balloon only, aspirin; stent, aspirin/coumadin/dipyridamole vs. aspirin/ticlopidine). Blood samples were taken at baseline and up to 7 days after PCI and PAI-1 active antigen and tissue plasminogen activator (t-PA) antigen were determined. Restenosis was angiographically determined after 6 months. RESULTS PCI increased both t-PA and PAI-1 levels (P < 0.001), with a significant prolonged and pronounced increase in stent vs. balloon-only patients (P < 0.05). Restenosis (stent 26%; balloon 38%) was significantly correlated to an attenuated PAI-1 increase after 24 h in the ticlopidine group (P = 0.007; restenosis, relative Delta PAI-1 + 50 +/- 28%; non-restenosis, + 139 +/- 50%), but not in the coumadin group. In the balloon-only group late restenosis (ISR) was associated with a trend for an augmented PAI-1 increase after 24 h. CONCLUSIONS Coronary stent implantation significantly increases t-PA and PAI-1 plasma levels up to 1 week compared with balloon angioplasty alone. ISR in ticlopidine-treated patients was associated with an attenuated early PAI-1 active antigen increase. A less than 50% increase 24 h after stent implantation under ticlopidine treatment may identify patients at risk for the development of ISR.
Collapse
Affiliation(s)
- G Christ
- Department of Cardiology, Medical University of Vienna, Währingergürtel 18-20, A-1090 Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Bochaton-Piallat ML, Gabbiani G. Modulation of smooth muscle cell proliferation and migration: role of smooth muscle cell heterogeneity. Handb Exp Pharmacol 2005:645-63. [PMID: 16596818 DOI: 10.1007/3-540-27661-0_24] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Proliferation and migration of smooth muscle cells (SMCs) from the media towards the intima are key events in atherosclerosis and restenosis. During these processes, SMC undergo phenotypic modulations leading to SMC dedifferentiation. The identification and characterization of factors controlling these phenotypic changes are crucial in order to prevent the formation of intimal thickening. One of the questions which presently remains open, is to know whether any SMCs of the media are capable of accumulating into the intima or whether only a predisposed medial SMC subpopulation is involved in this process. The latter hypothesis implies that arterial SMCs are phenotypically heterogenous. In this chapter, we will describe the distinct SMC phenotypes identified in arteries of various species, including humans. Their role in the formation of intimal thickening will be discussed.
Collapse
|
17
|
Providence KM, Higgins PJ. PAI-1 expression is required for epithelial cell migration in two distinct phases of in vitro wound repair. J Cell Physiol 2004; 200:297-308. [PMID: 15174100 DOI: 10.1002/jcp.20016] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Several proteases and their specific inhibitors modulate the interdependent processes of cell migration and matrix proteolysis as part of the global program of trauma repair. Expression of plasminogen activator inhibitor type-1 (PAI-1), a serine protease inhibitor (SERPIN) important in the control of barrier proteolysis and cell-to-matrix adhesion, for example, is spatially-temporally regulated following epithelial denudation injury in vitro as well as in vivo. PAI-1 mRNA/protein synthesis was induced early after epidermal monolayer scraping and restricted to keratinocytes comprising the motile cohort closely recapitulating, thereby, similar events during cutaneous healing. The time course of PAI-1 promoter-driven PAI-1-GFP fusion "reporter" expression in wound-juxtaposed cells approximated that of the endogenous PAI-1 gene confirming the location-specificity of gene regulation in this model. ERK activation was evident within 5 min after injury and particularly prominent in cells residing at the scrape-edge (suggesting a possible role in PAI-1 induction and/or the motile response) as was myosin light chain (MLC) phosphorylation. Indeed, MEK blockade with PD98059 or U0126 attenuated keratinocyte migration (by > or =60%), as did transient transfection of a dominant-negative ERK1 construct (40% decrease in monolayer repair), and completely inhibited PAI-1 transcript expression. Anti-sense down-regulation of PAI-1 synthesis (by 80-85%), or addition of PAI-1 neutralizing antibodies also inhibited injury site closure over a 24 h period establishing that PAI-1 was required for efficient long-term planar motility in this system. PAI-1 anti-sense transfection or actinomycin D transcriptional blockade, in contrast, did not affect the initial migratory response suggesting that residual PAI-1 protein levels (at least in transfectant cells and actinomycin D-treated cultures) may be sufficient to support early cell movement. Pharmacologic inhibition of keratinocyte MEK signaling effectively ablated scrape-induced PAI-1 mRNA expression but failed to attenuate wound-associated increases in cellular PAI-1 protein levels soon after monolayer injury. Collectively, these data suggest that basal PAI-1 transcripts may be mobilized for initial PAI-1 synthesis and, perhaps, the early motile response while maintenance of the normal rate of migration requires the prolonged PAI-1 expression that typically accompanies the repair response. To assess this possibility, scrape site closure studies were designed using keratinocytes isolated from PAI-1-/- mice. PAI-1-/- keratinocytes, in fact, had a significant wound healing defect evident even within the first 6 h following monolayer denudation injury. Addition of active PAI-1 protein to PAI-/- keratinocytes rescued the migratory phenotype that that approximating wild-type cells. These findings validate use of the present keratinocyte model to investigate injury-related controls on PAI-1 gene regulation and, collectively, implicate participation of PAI-1 in two distinct phases of epidermal wound repair.
Collapse
Affiliation(s)
- Kirwin M Providence
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, New York 12208, USA
| | | |
Collapse
|
18
|
Beyersdorf F. Myocardial and endothelial protection for heart transplantation in the new millenium: lessons learned and future directions. J Heart Lung Transplant 2004; 23:657-65. [PMID: 15366424 DOI: 10.1016/j.healun.2003.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Friedhelm Beyersdorf
- Department of Cardiovascular Surgery, Albert-Ludwigs-University Freiburg, Freiburg, Germany.
| |
Collapse
|
19
|
Dai E, Guan H, Liu L, Little S, McFadden G, Vaziri S, Cao H, Ivanova IA, Bocksch L, Lucas A. Serp-1, a viral anti-inflammatory serpin, regulates cellular serine proteinase and serpin responses to vascular injury. J Biol Chem 2003; 278:18563-72. [PMID: 12637546 DOI: 10.1074/jbc.m209683200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Complex DNA viruses have tapped into cellular serpin responses that act as key regulatory steps in coagulation and inflammatory cascades. Serp-1 is one such viral serpin that effectively protects virus-infected tissues from host inflammatory responses. When given as purified protein, Serp-1 markedly inhibits vascular monocyte invasion and plaque growth in animal models. We have investigated mechanisms of viral serpin inhibition of vascular inflammatory responses. In vascular injury models, Serp-1 altered early cellular plasminogen activator (tissue plasminogen activator), inhibitor (PAI-1), and receptor (urokinase-type plasminogen activator) expression (p < 0.01). Serp-1, but not a reactive center loop mutant, up-regulated PAI-1 serpin expression in human endothelial cells. Treatment of endothelial cells with antibody to urokinase-type plasminogen activator and vitronectin blocked Serp-1-induced changes. Significantly, Serp-1 blocked intimal hyperplasia (p < 0.0001) after aortic allograft transplant (p < 0.0001) in PAI-1-deficient mice. Serp-1 also blocked plaque growth after aortic isograft transplant and after wire-induced injury (p < 0.05) in PAI-1-deficient mice indicating that increase in PAI-1 expression is not required for Serp-1 to block vasculopathy development. Serp-1 did not inhibit plaque growth in uPAR-deficient mice after aortic allograft transplant. We conclude that the poxviral serpin, Serp-1, attenuates vascular inflammatory responses to injury through a pathway mediated by native uPA receptors and vitronectin.
Collapse
Affiliation(s)
- Erbin Dai
- Vascular Biology Research Group, John P. Robarts' Research Institute, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Benza RL, Anderson PG, Lyle K, Barchue J, de Oliveira AL, Cavender MA, Pinderski LJ, George JF. Donor PAI-1 expression inhibits the intimal response of early allograft vascular disease. J Heart Lung Transplant 2003; 22:515-8. [PMID: 12742412 DOI: 10.1016/s1053-2498(02)00662-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND The development of allograft vascular disease (AVD) may be related to altered expression of the fibrinolytic system. We determined the extent to which plasminogen activator inhibitor type 1 (PAI-1) expression in donor tissue influences intimal proliferation (IP) in a mouse model of AVD. METHODS We utilized an end-to-end abdominal aortic transplant model in mice to investigate the development of IP in 3 groups of 6 recipients. Group A (negative control) utilized C57BL/6J strain mice as both donors and recipients. In Groups B (positive control) and C, C57BL/6J mice were vessel donors and CBA/J mice were recipients. Both groups received intraperitoneal anti-CD4 and anti-CD8 monoclonal antibodies (250 microg/week for 5 weeks). Group C recipients, however, were transplanted with vessels from C57BL/6J PAI-1 knockout mice. Animals were killed at 50 days. Transplanted aortas were removed and intimal areas calculated using morphometric analysis. RESULTS Group A (mean intimal area 6421 +/- 8507 microm(2)) demonstrated very little IP in comparison to the other groups. IP was significantly higher in Group B (mean intimal area 56357 +/- 35629 microm(2)) than Group A (p = 0.008). Group C (mean intimal area 288195 +/- 123279 microm(2)) demonstrated significantly more intimal proliferation than either Groups A or B (vs B, p = 0.003; vs A, p < 0.001). The significance of these results is maintained if intimal thickness is measured as a stand-alone reference for the intimal response. CONCLUSIONS Lack of PAI-1 expression in donor tissue greatly exaggerates the extent of IP after allogeneic transplantation and suggests that PAI-1 is important in limiting the early phase of AVD.
Collapse
Affiliation(s)
- Raymond L Benza
- Division of Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Houard X, Monnot C, Dive V, Corvol P, Pagano M. Vascular smooth muscle cells efficiently activate a new proteinase cascade involving plasminogen and fibronectin. J Cell Biochem 2003; 88:1188-201. [PMID: 12647301 DOI: 10.1002/jcb.10460] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The plasminogen/plasmin system is involved in vascular wall remodeling after injury, through extracellular matrix (ECM) degradation and proteinase activation. Vascular smooth muscle cells (VSMCs) synthesize various components of the plasminogen/plasmin system. We investigated the conversion of plasminogen into plasmin in primary cultured rat VSMCs. VSMCs efficiently converted exogenous plasminogen into plasmin in a time- and dose-dependent manner. We measured plasmin activity by monitoring the hydrolysis of Tosyl-G-P-R-Mca, a fluorogenic substrate of plasmin. Cell-mediated plasmin activation was associated with the degradation of ECM, as revealed by fibronectin proteolysis. Plasmin also activated a proteinase able to hydrolyze Mca-P-L-G-L-Dpa-A-R-NH(2), a fluorogenic substrate of matrix metalloproteinases (MMPs). However, this proteinase was not inhibited by an MMP inhibitor. Furthermore, this proteinase displayed similar biochemical and pharmacological properties to fibronectin-proteinase, a recently identified zinc-dependent metalloproteinase located in the gelatin-binding domain of fibronectin. These results show that VSMCs convert exogenous plasminogen into plasmin in their pericellular environment. By hydrolyzing matrix protein plasmin activates a latent metalloproteinase that differs from MMP, fibronectin-proteinase. This metalloproteinase may participate to vascular wall remodeling, in concert with other proteinases.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic
- Enzyme Activation/drug effects
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Extracellular Matrix Proteins/metabolism
- Fibrinolysin/metabolism
- Fibronectins/biosynthesis
- Matrix Metalloproteinases/metabolism
- Muscle, Smooth, Vascular
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Plasminogen/biosynthesis
- Plasminogen/pharmacology
- Polymerase Chain Reaction
- RNA, Messenger/analysis
- RNA, Messenger/biosynthesis
- Rats
- Rats, Wistar
- Sequence Analysis, DNA
- Transcription, Genetic
- Wound Healing/physiology
Collapse
Affiliation(s)
- Xavier Houard
- INSERM Unité 36, Collège de France, 11 place Marcelin Berthelot, 75231 Paris Cedex 05, France
| | | | | | | | | |
Collapse
|
22
|
Askari AT, Brennan ML, Zhou X, Drinko J, Morehead A, Thomas JD, Topol EJ, Hazen SL, Penn MS. Myeloperoxidase and plasminogen activator inhibitor 1 play a central role in ventricular remodeling after myocardial infarction. J Exp Med 2003; 197:615-24. [PMID: 12615902 PMCID: PMC2193831 DOI: 10.1084/jem.20021426] [Citation(s) in RCA: 181] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Left ventricular (LV) remodeling after myocardial infarction (MI) results in LV dilation, a major cause of congestive heart failure and sudden cardiac death. Ischemic injury and the ensuing inflammatory response participate in LV remodeling, leading to myocardial rupture and LV dilation. Myeloperoxidase (MPO), which accumulates in the infarct zone, is released from neutrophils and monocytes leading to the formation of reactive chlorinating species capable of oxidizing proteins and altering biological function. We studied acute myocardial infarction (AMI) in a chronic coronary artery ligation model in MPO null mice (MPO(-/-)). MPO(-/-) demonstrated decreased leukocyte infiltration, significant reduction in LV dilation, and marked preservation of LV function. The mechanism appears to be due to decreased oxidative inactivation of plasminogen activator inhibitor 1 (PAI-1) in the MPO(-/-), leading to decreased tissue plasmin activity. MPO and PAI-1 are shown to have a critical role in the LV response immediately after MI, as demonstrated by markedly delayed myocardial rupture in the MPO(-/-) and accelerated rupture in the PAI-1(-/-). These data offer a mechanistic link between inflammation and LV remodeling by demonstrating a heretofore unrecognized role for MPO and PAI-1 in orchestrating the myocardial response to AMI.
Collapse
Affiliation(s)
- Arman T Askari
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Benza RL. Commentary: non-immunologic vascular failure of the transplanted heart. J Heart Lung Transplant 2003; 22:241-3. [PMID: 12633689 DOI: 10.1016/s1053-2498(02)00673-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Raymond L Benza
- University of Alabama at Birmingham, Department of Medicine, 1900 University Boulevard, 321 THT, Birmingham, Alabama 35294-0006, USA.
| |
Collapse
|
24
|
Cai WJ, Koltai S, Kocsis E, Scholz D, Kostin S, Luo X, Schaper W, Schaper J. Remodeling of the adventitia during coronary arteriogenesis. Am J Physiol Heart Circ Physiol 2003; 284:H31-40. [PMID: 12388238 DOI: 10.1152/ajpheart.00478.2002] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We studied the role of the adventitia in adaptive arteriogenesis during the phase of active growth of coronary collateral vessels (CV) induced by chronic occlusion of the left circumflex coronary artery in canine hearts. We used electron microscopy and immunoconfocal (IF) labeling for bFGF, matrix metalloproteinase (MMP)-2, MMP-9, tissue-type plasminogen activator (tPA), its inhibitor (PAI-1), fibronectin (FN), and Ki-67. Proliferation of smooth muscle cells and adventitial fibroblasts was evident. Quantitative IF showed that adventitial MMP-2, MMP-9, and FN were 9.2-, 7.5-, and 8.6-fold, bFGF was 5.1-fold, and PAI-1 was 3.4-fold higher in CV than in normal vessels (NV). The number of fibroblasts was 5-fold elevated in CV, but the elastic fiber content was 25-fold greater in NV than in CV. Perivascular myocyte damage and induction of endothelial nitric oxide synthase in peri-CV capillaries indicate expansion of CV. It was concluded that adventitial activation is associated with the development of CV through cell proliferation, production of growth factors, and induction of extracellular proteolysis thereby contributing to remodeling during adaptive arteriogenesis.
Collapse
Affiliation(s)
- Wei-Jun Cai
- Max Planck Institute, Department of Experimental Cardiology, D-61231 Bad Nauheim, Germany
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Botella LM, Sánchez-Elsner T, Sanz-Rodriguez F, Kojima S, Shimada J, Guerrero-Esteo M, Cooreman MP, Ratziu V, Langa C, Vary CPH, Ramirez JR, Friedman S, Bernabéu C. Transcriptional activation of endoglin and transforming growth factor-beta signaling components by cooperative interaction between Sp1 and KLF6: their potential role in the response to vascular injury. Blood 2002; 100:4001-10. [PMID: 12433697 DOI: 10.1182/blood.v100.12.4001] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Endoglin is an endothelial membrane glycoprotein involved in cardiovascular morphogenesis and vascular remodeling. It associates with transforming growth factor-beta (TGF-beta) signaling receptors to bind TGF-beta family members, forming a functional receptor complex. Arterial injury leads to up-regulation of endoglin, but the underlying regulatory events are unknown. The transcription factor KLF6, an immediate-early response gene induced in endothelial cells during vascular injury, transactivates TGF-beta, TGF-beta signaling receptors, and TGF-beta-stimulated genes. KLF6 and, subsequently, endoglin were colocalized to vascular endothelium (ie, expressed in the same cell type) following carotid balloon injury in rats. After endothelial denudation, KLF6 was induced and translocated to the nucleus; this was followed 6 hours later by increased endoglin expression. Transient overexpression of KLF6, but not Egr-1, stimulated endogenous endoglin mRNA and transactivated the endoglin promoter. This transactivation was dependent on a GC-rich tract required for basal activity of the endoglin promoter driven by the related GC box binding protein, Sp1. In cells lacking Sp1 and KLF6, transfected KLF6 and Sp1 cooperatively transactivated the endoglin promoter and those of collagen alpha1(I), urokinase-type plasminogen activator, TGF-beta1, and TGF-beta receptor type 1. Direct physical interaction between Sp1 and KLF6 was documented by coimmunoprecipitation, pull-down experiments, and the GAL4 one-hybrid system, mapping the KLF6 interaction to the C-terminal domain of Sp1. These data provide evidence that injury-induced KLF6 and preexisting Sp1 may cooperate in regulating the expression of endoglin and related members of the TGF-beta signaling complex in vascular repair.
Collapse
MESH Headings
- Animals
- Antigens, CD
- Carotid Arteries
- Catheterization/adverse effects
- Disease Models, Animal
- Drug Synergism
- Endoglin
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/injuries
- Endothelium, Vascular/metabolism
- Humans
- Kruppel-Like Factor 6
- Kruppel-Like Transcription Factors
- Promoter Regions, Genetic/drug effects
- Protein Binding
- Proto-Oncogene Proteins
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Cell Surface
- Signal Transduction
- Sp1 Transcription Factor/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Trans-Activators/pharmacology
- Transcriptional Activation
- Transfection
- Transforming Growth Factor beta/metabolism
- Umbilical Veins
- Vascular Cell Adhesion Molecule-1/genetics
- Vascular Cell Adhesion Molecule-1/metabolism
Collapse
Affiliation(s)
- Luisa M Botella
- Centro de Investigaciones Biológicas, Consejo Superior Investigaciones Cientificas (CSIC), Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Furman C, Luo Z, Walsh K, Duverger N, Copin C, Fruchart JC, Rouis M. Systemic tissue inhibitor of metalloproteinase-1 gene delivery reduces neointimal hyperplasia in balloon-injured rat carotid artery. FEBS Lett 2002; 531:122-6. [PMID: 12417298 DOI: 10.1016/s0014-5793(02)03388-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Metalloproteinases (MMP)-2 and MMP-9 play a role in smooth muscle cell (SMC) migration from the media to the intima following arterial injury. Intravenous administration of adenovirus encoding tissue inhibitor of metalloproteinase-1 (TIMP-1) into balloon-injured rat arteries (3 x 10(11) viral particles/rat; n=7) resulted in a transient expression of TIMP-1 and a significant inhibition of neointima thickening within 16 days ( approximately 40% vs. control; P=0.012). Three days after injury, the number of intimal SMCs was decreased by approximately 98% in TIMP-1-treated rats. However, no alteration was seen in intimal SMC proliferation after 13 days of injury. Therefore, our results show that systemic gene transfer of TIMP-1 is a promising approach in early restenosis treatment.
Collapse
Affiliation(s)
- C Furman
- Institut Pasteur de Lille and INSERM U-545, 1, rue du Professeur Calmette, Lille, France
| | | | | | | | | | | | | |
Collapse
|
27
|
Lau HKF, Ho J. Regulation of plasminogen activator inhibitor-1 secretion by growth factors in smooth muscle cells. Blood Coagul Fibrinolysis 2002; 13:541-9. [PMID: 12192306 DOI: 10.1097/00001721-200209000-00009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epithelioid-type vascular smooth muscle cells are metabolically active and secrete many proteases and protease inhibitors. We have previously cloned epithelioid-type smooth muscle cells from rat carotid arteries, and showed that polypeptide growth factors basic fibroblast growth factor (bFGF) and platelet-derived growth factor (PDGF) could dose-dependently induce plasminogen activator inhibitor-1 (PAI-1) secretion from these cells. In the present study, we have used these cells to investigate the growth factor-induced signal transduction pathways leading to PAI-1 secretion. We report here that PAI-1 induction was dependent on protein kinase C (PKC) and tyrosine kinase but not on protein kinase A (PKA), ras and phosphoinositol-3-kinase inhibitor. Induction of PAI-1 by bFGF and PDGF was also accompanied by activation of a mitogen-activated protein kinase pathway involving Raf/Mek/Erk1/2, and the family non-receptor tyrosine kinases., another non-receptor tyrosine kinase, on the contrary, behaved differently from in that it was part of a pathway leading to PAI-1 induction by bFGF, but not when PDGF was used as the stimulating reagent. Activation of a PKA-dependent pathway(s) opposed PAI-1 induction. One mechanism for PKA activators to inhibit PAI-1 secretion was that they markedly inhibited the phosphorylations of Mek and mitogen-activated protein kinase that were up-regulated in the presence of bFGF and PDGF.
Collapse
Affiliation(s)
- H K F Lau
- Division of Hematology, Department of Medicine, St Michael's and University of Toronto, Ontario, Canada.
| | | |
Collapse
|
28
|
Dovjak N, Blinc A, Keber D. In vivo release of tissue-type plasminogen activator antigen from the human brachial artery. Thromb Res 2002; 106:249-55. [PMID: 12297133 DOI: 10.1016/s0049-3848(02)00132-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The rate of secretion of t-PA from vascular endothelial cells has been proposed as a good marker of endothelial function. Our aim was to measure the rate of in vivo t-PA antigen release from the human brachial artery and not from the combined vascular pool of the upper extremity. In 10 healthy male volunteers, we have occluded the forearm by a sphygmomanometer cuff for 5-7 min in order to reduce the blood flow in the brachial artery. Arterial blood samples were taken from the cubital artery above the occlusion and from the contralateral artery that served as the control. The arterial t-PA antigen concentrations were significantly higher after forearm occlusion than in the non-occluded contralateral arteries: median 3.3 (range between first and third quartile 2.1-3.6) vs. 2.5 (2.0-3.2) ng/ml, p=0.03. The PAI-1 antigen concentrations did not change significantly: 6.9 (2.3-18.6) ng/ml in the contralateral artery vs. 5.4 (1.8-8.0) ng/ml after occlusion, p=0.09. The average forward blood flow in the distal 15 cm of the brachial artery that was measured by duplex ultrasound decreased from 107 (97-118) ml/min at baseline to 25 ml/min (19-33) ml/min during occlusion. The release rate of circulating t-PA antigen was calculated as the product of the increment in arterial t-PA concentration and the average plasma flow in the arterial segment of interest with a volume of 2 ml. The median release rate of t-PA antigen under conditions of reduced blood flow in the brachial artery was 3.3 (1.1-11.5) ng/min. We conclude that the secretion of t-PA antigen from arterial endothelium of healthy subjects is substantial, whereas the arterial wall is not an important source of PAI-1 in vivo.
Collapse
Affiliation(s)
- Neda Dovjak
- Department of Vascular Diseases, University of Ljubljana Medical Center, Riharjeva 24, SI-1000 Ljubljana, Slovenia
| | | | | |
Collapse
|
29
|
Watanabe A, Kanai H, Arai M, Sekiguchi K, Uchiyama T, Nagai R, Kurabayashi M. Retinoids induce the PAI-1 gene expression through tyrosine kinase-dependent pathways in vascular smooth muscle cells. J Cardiovasc Pharmacol 2002; 39:503-12. [PMID: 11904524 DOI: 10.1097/00005344-200204000-00006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Retinoids exert their pleiotropic effects on several pathophysiologic processes, including neointima formation after experimental vascular injury. Plasminogen activator inhibitor-1 (PAI-1) has been proposed to play an inhibitory role in arterial neointima formation after injury. We examined whether retinoids regulate PAI-1 expression in cultured vascular smooth muscle cells (SMCs). Northern blot analysis showed that all-trans retinoic acid (atRA) and 9-cis retinoic acid (9cRA) increased PAI-1 mRNA levels in a dose-dependent manner. These responses were completely inhibited by tyrosine kinase inhibitors. The half-life of PAI-1 was not affected by atRA, suggesting that induction of PAI-1 mRNA was mainly regulated at the transcriptional levels. Stable and transient transfection assays of the human PAI-1 promoter-luciferase constructs indicate that DNA sequence responsive to either ligand-stimulated or overexpressed retinoic acid receptor-alpha expression vector lies downstream of -363 relative to the transcription start site, where no putative retinoic acid response element is found. These results indicate that atRA and 9cRA increase PAI-1 gene transcription through pathways involving tyrosine kinases in SMCs. Because PAI-1 inhibits the production of fibrinolytic protein plasmin that facilitates SMC migration, induction of the PAI-1 gene expression by atRA may at least partly account for the role of atRA as an important inhibitor of neointima formation.
Collapse
Affiliation(s)
- Atai Watanabe
- Second Department of Internal Medicine, Gunma University School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | | | | | | | | | | | | |
Collapse
|
30
|
Plekhanova O, Parfyonova Y, Bibilashvily R, Domogatskii S, Stepanova V, Gulba DC, Agrotis A, Bobik A, Tkachuk V. Urokinase plasminogen activator augments cell proliferation and neointima formation in injured arteries via proteolytic mechanisms. Atherosclerosis 2001; 159:297-306. [PMID: 11730809 DOI: 10.1016/s0021-9150(01)00511-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Urokinase plasminogen activator (uPA) has been implicated in the healing responses of injured arteries, but the importance of its various properties that influence smooth muscle cell (SMC) proliferation and migration in vivo is unclear. We used three recombinant (r-) forms of uPA, which differ markedly in their proteolytic activities and abilities to bind to the uPA receptor (uPAR), to determine, which property most influences the healing responses of balloon catheter injured rat carotid arteries. After injury, uPA and uPAR expression increased markedly throughout the period when medial SMCs were rapidly proliferating and migrating to form the neointima. Perivascular application of uPA neutralizing antibodies immediately after injury attenuated the healing response, significantly reducing neointima size and neointimal SMC numbers. Perivascular application of r-uPAwt (wild type uPA) or r-uPA/GDF (r-uPA with multiple mutations in its growth factor-like domain) doubled the size of the neointima. Four days after injury these two uPAs nearly doubled neointimal and medial SMC numbers in the vessels, and induced greater reductions in lumen size than injury alone. Proteolytically inactive r-uPA/H/Q (containing glutamine rather than histidine-204 in its catalytic site) did not affect neointima or lumen size. Also, in contrast to the actions of proteolytically active uPAs, tissue plasminogen activator (tPA) did not affect the rate of neointima development. We conclude that uPA is an important factor regulating the healing responses of balloon catheter injured arteries, and its proteolytic property, which cannot be mimicked by tPA, greatly influences SMC proliferation and early neointima formation.
Collapse
Affiliation(s)
- O Plekhanova
- Molecular Endocrinology Laboratory, Institute of Experimental Cardiology, Cardiology Research Center, 121552, Moscow, Russia
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ortlepp JR, Hoffmann R, Killian A, Lauscher J, Merkelbach-Brese S, Hanrath P. The 4G/5G promotor polymorphism of the plasminogen activator inhibitor-1 gene and late lumen loss after coronary stent placement in smoking and nonsmoking patients. Clin Cardiol 2001; 24:585-91. [PMID: 11558839 PMCID: PMC6654891 DOI: 10.1002/clc.4960240904] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2000] [Accepted: 11/13/2000] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Instent restenosis remains a significant clinical problem. Identification of patients at risk for instent restenosis may allow selection of individualized appropriate therapeutic approaches. Genetic polymorphisms have been suggested to be associated with the risk of instent restenosis. Smoking is known to influence hemostatic parameters. HYPOTHESIS This study investigated the influence of the 4G/5G promotor polymorphism of the plasminogen activator inhibitor type I (PAI-1) gene on instent restenosis in smoking and nonsmoking patients. METHODS In all, 300 consecutive patients (133 nonsmoking; 167 smoking) with elective coronary stent placement and 6-month angiographic follow-up were studied. Quantitative coronary angiography and genotyping with polymerase chain reaction analysis were performed in all patients. RESULTS Nonsmoking PAI-1 4G/4G carriers showed a significantly greater late lumen loss (n = 38; 0.54 +/- 0.53 mm) compared with nonsmoking PAI-1 4G/5G (n = 68; 0.38 +/- 0.45 mm) or 5G/5G (n = 27; 0.19 +/- 0.23 mm) carriers, analysis of variance (ANOVA) p < 0.001. Smoking patients with the genotypes 4G/4G (n = 46; 0.53 +/- 0.54 mm) and 4G/5G (n = 79; 0.37 +/- 0.41 mm) had a late loss similar to that of nonsmoking patients. Smoking 5G/5G carriers had the highest late loss of all smoking patients (n = 42; 0.63 +/- 0.50); ANOVA p < 0.05; nonsmoking 5G/5G vs. smoking 5G/5G p < 0.001. CONCLUSION The promotor polymorphism of the PAI-1 gene has a significant influence on instent restenosis after coronary stent implantation. The 5G/5G genotype predisposes nonsmoking gene carriers to less late lumen loss, whereas in smoking gene carriers this genotype is associated with the greatest late lumen loss. This might be explained by an altered expression pattern of hemostatic parameters.
Collapse
Affiliation(s)
- J R Ortlepp
- Medical Clinic I, University Hospital of Aachen, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Coutts A, Chen G, Stephens N, Hirst S, Douglas D, Eichholtz T, Khalil N. Release of biologically active TGF-beta from airway smooth muscle cells induces autocrine synthesis of collagen. Am J Physiol Lung Cell Mol Physiol 2001; 280:L999-1008. [PMID: 11290525 DOI: 10.1152/ajplung.2001.280.5.l999] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In severe or chronic asthma, there is an increase in airway smooth muscle cell (ASMC) mass as well as an increase in connective tissue proteins in the smooth muscle layer of airways. Transforming growth factor-beta (TGF-beta) exists in three isoforms in mammals and is a potent regulator of connective tissue protein synthesis. Using immunohistochemistry, we had previously demonstrated that ASMCs contain large quantities of TGF-beta1-3. In this study, we demonstrate that bovine ASMC-derived TGF-beta associates with the TGF-beta latency binding protein-1 (LTBP-1) expressed by the same cells. The TGF-beta associated with LTBP-1 localizes TGF-beta extracellularly. Furthermore, plasmin, a serine protease, regulates the secretion of a biologically active form of TGF-beta by ASMCs as well as the release of extracellular TGF-beta. The biologically active TGF-beta released by plasmin induces ASMCs to synthesize collagen I in an autocrine manner. The autocrine induction of collagen expression by ASMCs may contribute to the irreversible fibrosis and remodeling seen in the airways of some asthmatics.
Collapse
Affiliation(s)
- A Coutts
- Department of Medicine and Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, Manitoba R3E 0V9
| | | | | | | | | | | | | |
Collapse
|
33
|
Lamfers ML, Lardenoye JH, de Vries MR, Aalders MC, Engelse MA, Grimbergen JM, van Hinsbergh VW, Quax PH. In vivo suppression of restenosis in balloon-injured rat carotid artery by adenovirus-mediated gene transfer of the cell surface-directed plasmin inhibitor ATF.BPTI. Gene Ther 2001; 8:534-41. [PMID: 11319620 DOI: 10.1038/sj.gt.3301437] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2000] [Accepted: 01/09/2001] [Indexed: 11/09/2022]
Abstract
Injury-induced neointimal development results from migration and proliferation of vascular smooth muscle cells (SMC). Cell migration requires controlled proteolytic degradation of extracellular matrix surrounding the cell. Plasmin is a major contributor to this process by degrading various matrix proteins directly, or indirectly by activating matrix metalloproteinases. This makes it an attractive target for inhibition by gene transfer. An adenoviral vector, Ad.ATF.BPTI, was constructed encoding a hybrid protein, which consists of the aminoterminal fragment (ATF) of urokinase-type plasminogen activator (u-PA) linked to bovine pancreas trypsin inhibitor (BPTI), a potent inhibitor of plasmin. This hybrid protein binds to the u-PA receptor, thereby inhibiting plasmin activity at the cell surface, and was found to be a potent inhibitor of cell migration in vitro. Local infection with Ad.ATF.BPTI of balloon-injured rat carotid artery resulted in detectable expression of ATF.BPTI mRNA and protein in the vessel wall. Morphometric analysis of arterial cross-sections revealed that delivery of Ad.ATF.BPTI to the carotid artery wall at the time of balloon injury inhibited neointima formation by 53% (P < 0.01) at 14 days and 19% (P = NS) at 28 days after injury when compared with control vector-infected arteries. Intima/media ratios were decreased by 60% (P < 0.01) and 35% (P < 0.05) at 14 and 28 days, respectively, when compared with control vector-infected arteries. Furthermore, a small but significant increase in medial area was found in the Ad.ATF.BPTI-treated arteries at 28 days (P < 0.05). These results show that local infection of the vessel wall with Ad.ATF.BPTI reduces neointima formation, presumably by inhibiting SMC migration, thereby offering a novel therapeutic approach to inhibiting neointima development.
Collapse
Affiliation(s)
- M L Lamfers
- Gaubius Laboratory TNO-PG, 2301 CE Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kutz SM, Providence KM, Higgins PJ. Antisense targeting of c-fos transcripts inhibits serum- and TGF-beta 1-stimulated PAI-1 gene expression and directed motility in renal epithelial cells. CELL MOTILITY AND THE CYTOSKELETON 2001; 48:163-74. [PMID: 11223948 DOI: 10.1002/1097-0169(200103)48:3<163::aid-cm1006>3.0.co;2-c] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Plasminogen activator inhibitor type-1 (PAI-1), the major regulator of pericellular plasmin generation, and the c-FOS transcription factor are expressed by migrating cells in response to monolayer wounding. Induced c-fos and PAI-1 transcripts were evident within 30 min and 2 h, respectively, of scrape injury to confluent, growth-arrested, cultures of NRK epithelial cells. Since c-FOS/AP-1 DNA-binding activity modulates both basal and inducible modes of PAI-1 gene control, and AP-1 motif binding factors were present in quiescent as well as stimulated NRK cells, a model of directionally regulated cell movement (migration into scrape-denuded "wounds") was used to assess the consequences of c-fos transcript targeting on PAI-1 expression and cell motility. This in vitro model of epithelial injury closely approximated in vivo wound repair with regard to the spatial and temporal emergence of cohorts of cells involved in migration, proliferation, and PAI-1 expression. Stable cell lines (NRKsof) were generated by transfection of parental NRK cells with a c-fos antisense expression vector. Serum-inducible c-fos transcripts and PAI-1 protein levels were significantly attenuated in NRKsof transfectants relative to parental controls or cells transfected with a neo(R) vector without the sof insert. NRKsof cells had a markedly impaired ability to repair scrape-generated monolayer wounds under basal, serum-stimulated, or TGF-beta 1-supplemented culture conditions. Since injury closure and PAI-1 induction were attenuated in c-fos antisense cells, it was important to clarify the role of specific AP-1 sites in serum-mediated PAI-1 transcription. PAI-1 "promoter"-driven CAT reporter expression was assessed within the real time of serum-stimulated PAI-1 induction. A segment of the PAI-1 promoter corresponding to nucleotides -533 to -764 upstream of the transcription start site functioned as a prominent serum-responsive region (SSR). The 9-fold increase in CAT mRNA levels attained with the -533 to -764 bp PAI-1 SRR ligated to a minimal PAI-1 promoter (i.e., 162 bp of 5' flanking sequence containing the basal transcription complex) closely approximated the serum-induced transcriptional activity of a fully responsive (1,230 bp) PAI-1 promoter construct as well as the endogenous PAI-1 gene. AP-1-like, CTF/NF-1-like, and AP-2 sites were identified in the SRR. The SRR AP-1 motif was homologous to the sequence TGACACA that mapped between nucleotides -740 and -703 in the human PAI-1 gene, a region essential for growth factor-inducible PAI-1 transcription. While the functionality of this AP-1 site in wound-regulated PAI-1 synthesis remains to be determined, antisense c-fos transcripts effectively attenuated PAI-1 induction and basal as well as growth factor-stimulated cell locomotion, indicating that expression of both the PAI-1 and c-fos genes is necessary for wound-initiated NRK cell migration.
Collapse
Affiliation(s)
- S M Kutz
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208, USA
| | | | | |
Collapse
|
35
|
Zablocki DK, Rade JJ, Alevriadou BR. Adenovirus-mediated expression of tissue plasminogen activator does not alter endothelial cell proliferation and invasion. In Vitro Cell Dev Biol Anim 2000; 36:625-8. [PMID: 11229592 DOI: 10.1290/1071-2690(2000)036<0625:ameotp>2.0.co;2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
36
|
Plekhanova OS, Parfyonova YV, Bibilashvily RS, Stepanova VV, Erne P, Bobik A, Tkachuk VA. Urokinase plasminogen activator enhances neointima growth and reduces lumen size in injured carotid arteries. J Hypertens 2000; 18:1065-9. [PMID: 10953998 DOI: 10.1097/00004872-200018080-00011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Increases in urokinase plasminogen activator (uPA) have been reported in tissues undergoing remodelling, but its effects on the vessel intima formation are not known. We investigated its effects on carotid artery intima, media and lumen size, as well as smooth muscle cell (SMC) proliferation and migration in vivo. DESIGN AND METHODS Carotid arteries of rats were distended with an inflated balloon catheter and uPA, or uPA-neutralizing antibodies were applied perivascularly in pluronic gel; control rats received vehicle. Carotid artery structure, cell migration and proliferation were assessed after 4 days by quantitative morphometry and immunohistochemistry. RESULTS Four days after increasing vessel uPA, the intima/media ratio was double compared to that in control rats (both P < 0.05). The size of the lumen reduced by 75%, compared to the vehicle-treated vessels (P < 0.05). The elevation in uPA also increased SMC numbers in the intima and media, compared to the vehicle-treated vessels (both P < 0.05). Antibody neutralizing endogenous uPA attenuated the growth responses in the distended arteries, reduced neointimal SMC numbers by approximately 50% and prevented much of the reduction in lumen size. CONCLUSIONS Thus, local increases in uPA in distended, injured arteries augment SMC migratory and proliferative responses, leading to increases in the thickness of the carotid artery intima and media and a reduction in lumen size; effects at least partially attributable to its proteolytic properties.
Collapse
|
37
|
Li F, Goncalves J, Faughnan K, Steiner MG, Pagan-Charry I, Esposito D, Chin B, Providence KM, Higgins PJ, Staiano-Coico L. Targeted inhibition of wound-induced PAI-1 expression alters migration and differentiation in human epidermal keratinocytes. Exp Cell Res 2000; 258:245-53. [PMID: 10896775 DOI: 10.1006/excr.2000.4918] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the adult epidermis, keratinocytes do not normally express the type-1 inhibitor of plasminogen activator (PAI-1). Basal epithelial cell-specific PAI-1 synthesis, however, accompanies epidermal wound repair in vivo in which PAI-1 transcripts and immunoreactive protein are confined to epithelial cells in the migrating tongue and the hyperproliferative zone. A model system using human keratinocytes (HaCaT cells) was developed to assess functional relationships between epithelial growth state transitions and PAI-1 expression. PAI-1 synthesis was maximal in low population density, exponentially growing HaCaT cultures; relative PAI-1 mRNA and protein levels progressively declined as cells attained, and were maintained in, a postconfluent condition. While the fraction of PAI-1(+) keratinocytes remained stable (at approximately 85-90% of the population) throughout the culture period, both PAI-1 mRNA abundance and mean cell-associated PAI-1 protein declined by >90% during prolonged (i.e., 8-day) growth arrest. Similar to epidermal trauma in vivo, scrape wounding of HaCaT monolayers resulted in the rapid and location-specific induction of PAI-1 protein (an increase of 11- to 16-fold relative to unwounded cultures) in cells immediately bordering the injury site. PAI-1 expression was evident in keratinocytes that comprised the opposed migrating fronts and remained elevated until wound closure. Down-regulation of PAI-1 synthesis in HaCaT cells transfected with an inducible LacSwitch-based antisense vector system markedly impaired both the rate and the extent of wound closure. All injuries created in antisense PAI-1 monolayers remained unhealed at day 8 postinjury compared to the 3-day complete repair typical of control cultures. Vector-driven modulation of PAI-1 synthesis was also associated with an increase in the percentage of suprabasal-type keratinocytes within the wound field. PAI-1 expression by migrating HaCaT cells appears necessary to maintain the basal epidermal phenotype and/or appropriate cell-to-substrate adhesion during injury repair.
Collapse
Affiliation(s)
- F Li
- Department of Surgery, Cornell University Medical College, 1300 York Avenue, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Busuttil SJ, Drumm C, Ploplis VA, Plow EF. Endoluminal arterial injury in plasminogen-deficient mice. J Surg Res 2000; 91:159-64. [PMID: 10839966 DOI: 10.1006/jsre.2000.5922] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Vascular remodeling following arterial injury is characterized by an initial inflammatory reaction. Prior experiments using peritoneal inflammatory models have shown that the plasminogen system plays a role in the intensity of the inflammatory response. This study was undertaken to test the hypothesis that an absence of plasminogen would lead to a decrease in vascular remodeling. METHODS A left carotid artery injury was created with a flexible guidewire in both wild-type [Plg(+/+)] and plasminogen deficient [Plg(-/-)] mice. The right carotid artery was uninjured and used as a control. Three weeks postinjury, the mice were sacrificed and perfusion fixed, and the bilateral carotid arteries were sectioned for histological examination and collection of morphometric data. RESULTS After arterial injury, electron microscopy of the acutely injured artery revealed that the endothelium was denuded, that there were breaks in the internal elastic membrane, and that there was disruption of the medial layer of smooth muscle cells. The intimal and medial areas were significantly increased between the uninjured and injured carotid arteries of both Plg(+/+) (+80% intimal, +41% medial, P < 0. 05) and Plg(-/-) [+48% intimal, +24% medial, P < 0.05) mice. However, although there was a significant increase in the adventitial area of Plg(+/+) mice (+18%, P < 0.05), there was no difference in Plg(-/-) mice (-6%). Interestingly, even after 3 weeks, four of six injured arteries in Plg(-/-) mice had persistent thrombus within the medial layer, whereas this was not found in any of the nine Plg(+/+) mouse arteries. DISCUSSION Plasminogen deficiency inhibited the increase in adventitial area seen after injury in Plg(+/+) mice, but not the increase in intimal or medial areas. Not surprisingly, plasminogen-deficient mice also demonstrated a severe alteration in intramural thrombus clearance. Thus, specific aspects of the vascular remodeling response are dependent on plasminogen.
Collapse
Affiliation(s)
- S J Busuttil
- Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
39
|
Cai W, Vosschulte R, Afsah-Hedjri A, Koltai S, Kocsis E, Scholz D, Kostin S, Schaper W, Schaper J. Altered balance between extracellular proteolysis and antiproteolysis is associated with adaptive coronary arteriogenesis. J Mol Cell Cardiol 2000; 32:997-1011. [PMID: 10888253 DOI: 10.1006/jmcc.2000.1137] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To study the role of extracellular proteolysis and antiproteolysis during adaptive arteriogenesis (collateral vessel growth) we took 58 collaterals at various developmental stages from 14 dogs with chronic occlusion of the left circumflex coronary artery (LCx) by ameroid constrictor. Immunofluorescence and quantitative immunofluorescence with antibodies against alpha-smooth muscle actin, desmin, matrix metalloproteinases 2 (MMP-2), MMP-9, tissue inhibitor of metalloproteinases 1 (TIMP-1) and 2 (TIMP-2), urokinase-type plasminogen activator (u-PA) and its inhibitor-1 (PAI-1) were studied with confocal microscopy. Additionally, SDS-PAGE zymography was employed. We found that in normal coronary arteries, MMP-2, MMP-9 and PAI-1 were present in all layers of the wall in small amounts. TIMP-1 was found only in smooth muscle cells. In contrast, in growing collaterals, MMP-2 and MMP-9 were 3.4-fold and 4.1-fold higher in the neointima than in the media respectively. TIMP-1 was 4.4-fold higher in the media over the growing neointima. Zymography showed MMP-2 and MMP-9 activated. PAI-1 was increased, especially in the growing neointima where it was 1.4-fold higher. In mature collaterals, MMP-2 and MMP-9 were downregulated in the neointima, 1.4-fold and 1.3-fold higher over the media. TIMP-1 was 1.4-fold increased in the neointima but PAI-1 was downregulated. Desmin and alpha-smooth muscle actin were significantly increased in the neointima compared to growing vessels. U-PA was moderately increased in growing vessels. TIMP-2 was not detectable in collaterals. We conclude that expression of MMP-2 and 9, TIMP-1 and PAI-1 showed a spatial and temporal pattern which is closely associated with the development of collateral vessels. The shift of the balance between proteolysis and antiproteolysis is regulated not only by MMPs and TIMP-1, but also by the PA-PAI system.
Collapse
Affiliation(s)
- W Cai
- Max-Planck-Institute, Dept. of Exp. Cardiology, Bad Nauheim, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lee ES, Bauer GE, Caldwell MP, Santilli SM. Association of artery wall hypoxia and cellular proliferation at a vascular anastomosis. J Surg Res 2000; 91:32-7. [PMID: 10816346 DOI: 10.1006/jsre.2000.5891] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND We hypothesize that arterial wall hypoxia incites the pathologic formation of intimal hyperplasia at an artery anastomosis. We have determined from previous studies performed in our laboratory, the oxygen tension profiles of the artery wall at various times after vascular anastomosis. The purpose of this study is to determine the rate of cellular proliferation at an artery anastomosis when the artery wall is most hypoxic. MATERIALS AND METHODS Expanded polytetrafluoroethylene (ePTFE) grafts were placed end to end in the infrarenal aorta of 27 New Zealand white rabbits. The anastomotic aortic wall oxygen (O(2)) tensions were measured with an O(2) microelectrode in rabbits 0, 7, 14, 28, and 42 days after surgery. O(2) tensions were also measured in 4 control rabbits for comparison. 5-Bromo-2'-deoxyuridine (BrDU) was injected intraperitoneally 24 h prior to rabbit sacrifice. After O(2) tension measurements, the rabbits were sacrificed and the aortic grafts were harvested. Bioquant morphometrics was used to measure cells with BrDU counterstaining and intimal thickness in 17 rabbits: in control (n = 4), Day 0 (n = 4), 7 (n = 5), and 42 (n = 4). Student's t test was used to compare O(2) tensions, cellular proliferation, and intimal hyperplasia between days. RESULTS The pO(2) levels at the outer layers of the aorta, 1 mm distal to the distal aortic graft anastomosis, were 61.0 +/- 2 (+/-SE) mm Hg for controls, 19.8 +/- 1 mm Hg for Day 7 (P < 0.0001), 19.0 +/- 1 mm Hg for Day 14, 39.2 +/- 1 mm Hg for Day 28, and 58.5 +/- 1 mm Hg for Day 42 aortic grafts. BrDU-staining ratios in the intima were significantly higher in the Day 7 aortic grafts, 28.6 +/- 3%, versus BrDU-staining ratio, 1.4 +/- 1%, in Day 42 aortic grafts (P < 0.0002). CONCLUSIONS Cellular proliferation is highest at Day 7 when the artery wall is most hypoxic and returns to baseline as O(2) tensions normalize.
Collapse
MESH Headings
- Anastomosis, Surgical
- Animals
- Aorta, Abdominal/chemistry
- Aorta, Abdominal/pathology
- Aorta, Abdominal/surgery
- Blood Vessel Prosthesis Implantation
- Bromodeoxyuridine/analysis
- Cell Division/physiology
- Hyperplasia
- Hypoxia/pathology
- Muscle, Smooth, Vascular/chemistry
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/surgery
- Oxygen Consumption
- Polytetrafluoroethylene
- Rabbits
- Tunica Intima/chemistry
- Tunica Intima/pathology
- Tunica Intima/surgery
Collapse
Affiliation(s)
- E S Lee
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
41
|
Bickerstaff AA, Xia D, Pelletier RP, Orosz CG. Mechanisms of graft acceptance: evidence that plasminogen activator controls donor-reactive delayed-type hypersensitivity responses in cardiac allograft acceptor mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:5132-9. [PMID: 10799871 DOI: 10.4049/jimmunol.164.10.5132] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have used delayed-type hypersensitivity (DTH) responses to probe the mechanisms of drug-induced cardiac allograft acceptance in mice. DBA/2-->C57BL/6 cardiac allograft recipients treated transiently with gallium nitrate accept their grafts for >90 days and fail to display DBA/2-reactive DTH responses. These DTH responses are restored when anti-TGF-beta Abs are included at the challenge site, and cell depletion studies showed that this DTH inhibition is mediated by CD4+ cells. Real-time PCR analysis revealed that allograft acceptor mice produce no more than background levels of TGF-beta mRNA at DTH challenge sites. This suggests that DTH regulation in allograft acceptor mice may involve TGF-beta activation, rather than TGF-beta production. The protease, plasmin, can activate TGF-beta, and activated T cells can express a receptor for the plasmin-producing enzyme urokinase-type plasminogen activator (uPA), and can also produce both uPA and tissue-type plasminogen activator (tPA). We observed that Abs to tPA or uPA can replace anti-TGF-beta mAb for the restoration of donor-reactive DTH responses in allograft acceptor mice. Histologic analysis revealed that accepted cardiac allografts express uPA, tPA, and active TGF-beta, whereas accepted cardiac isografts express only tPA, but not uPA or activated TGF-beta. These data demonstrate that local tPA and uPA contribute to DTH regulation in allograft acceptor mice and suggest that these elements of the fibrinolytic pathway are used to control donor-reactive cell-mediated immunity in allograft acceptor mice.
Collapse
Affiliation(s)
- A A Bickerstaff
- Department of Surgery, Comprehensive Cancer Center, Ohio State University College of Medicine, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
42
|
Plekhanova OS, Volynskaya EA, Kalinina NI, Parfenova EV. Urokinase stimulates differentiation of fibroblasts into myofibroblasts and their proliferation in damaged adventitia. Bull Exp Biol Med 2000; 129:429-32. [PMID: 10977941 DOI: 10.1007/bf02439792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/1999] [Indexed: 11/26/2022]
Abstract
Urokinase expression in the adventitia of rat common carotid artery increased on the 4th day after periadventitial damage. Periadventitial application of recombinant urokinase increased the area of the adventitia and the content of contractile and proliferating cells, while proteolytically inactive recombinant urokinase was ineffective.
Collapse
Affiliation(s)
- O S Plekhanova
- Russian Cardiological Research-and-Production Complex, Russian Ministry of Health, Moscow. plekhanova @mail.ru
| | | | | | | |
Collapse
|
43
|
Abstract
Since its introduction into clinical practice, more than 20 years ago, percutaneous transluminal coronary angioplasty (PTCA) has proven to be an effective, minimally invasive alternative to coronary artery bypass grafting (CABG). During this time there have been great improvements in the design of balloon catheters, operative procedures and adjuvant drug therapy, and this has resulted in low rates of primary failure and short-term complications. However, the potential benefits of angioplasty are diminished by the high rate of recurrent disease. Up to 40% of patients undergoing angioplasty develop clinically significant restenosis within a year of the procedure. Although the deployment of endovascular stents at the time of angioplasty improves the short-term outcome, 'in-stent' stenosis remains an enduring problem. In order to gain an insight into the mechanisms of restenosis, several experimental models of angioplasty have been developed. These have been used together with the tools provided by recent advances in molecular biology and catheter design to investigate restenosis in detail. It is now possible to deliver highly specific molecular antagonists, such as antisense gene sequences, to the site of injury. The knowledge provided by these studies may ultimately lead to novel forms of intervention. The present review is a synopsis of our current understanding of the pathological mechanisms of restenosis.
Collapse
Affiliation(s)
- G A Ferns
- Centre for Clinical Science and Measurement, School of Biological Sciences, University of Surrey, Guildford; The Royal Surrey County Hospital, Guildford, UK.
| | | |
Collapse
|
44
|
Molecular interactions between the plasminogen/plasmin and matrix metalloproteinase systems. ACTA ACUST UNITED AC 2000. [DOI: 10.1054/fipr.2000.0065] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
45
|
Providence KM, Kutz SM, Staiano-Coico L, Higgins PJ. PAI-1 gene expression is regionally induced in wounded epithelial cell monolayers and required for injury repair. J Cell Physiol 2000; 182:269-80. [PMID: 10623891 DOI: 10.1002/(sici)1097-4652(200002)182:2<269::aid-jcp16>3.0.co;2-e] [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: 01/15/2023]
Abstract
Induced expression of plasminogen activator inhibitor type-1 (PAI-1), a major negative regulator of pericellular plasmin generation, accompanies wound repair in vitro and in vivo. Since transcriptional control of the PAI-1 gene is superimposed on a growth state-dependent program of cell activation (Kutz et al., 1997, J Cell Physiol 170:8-18), it was important to define potentially functional relationships between PAI-1 synthesis and subpopulations of cells that emerge during the process of injury repair in T2 renal epithelial cells. Specific cohorts of migratory and proliferating cells induced in response to monolayer trauma were spatially as well as temporally distinct. Migrating cells did not divide in the initial 12 to 20 h postinjury. After 24 h, S-phase cells were generally restricted to a region 1 to 2 mm from, and parallel to, the wound edge. Proliferation of wound bed cells occurred subsequent to wound closure, whereas the distal contact-inhibited monolayer remained generally quiescent. Hydroxyurea blockade indicated, however, that proliferation (most likely of cells immediately behind the motile "tongue") was necessary for maintenance of cell-to-cell cohesiveness in the advancing front, although the ability to migrate was independent of proliferation. PAI-1 mRNA expression was rapidly up-regulated in response to wounding with inductive kinetics approximating that of serum-stimulated cultures. Differential harvesting of T2 cell subpopulations, based on proximity to the injury site, prior to Northern assessments of PAI-1 mRNA abundance indicated that PAI-1 transcripts were restricted to cells immediately bordering the wound or actively migrating and not expressed by cells in the distal contact-inhibited monolayer regions. Such cell location-specific distribution of PAI-1-producing cells was confirmed by immunocytochemistry. PAI-1 synthesis in cells that locomoted into the wound field continued until injury closure. Down-regulation of PAI-1 synthesis and matrix deposition in renal epithelial cells, stably transfected with a PAI-1 antisense expression vector, significantly impaired wound closure. Transfection of the wound repair-deficient R/A epithelial line with a sense PAI-1 expression construct restored both approximately normal levels of PAI-1 synthesis and repair ability. These data indicate that PAI-1 induction is an early event in creation of the wound-activated phenotype and appears to participate in the regulation of renal epithelial cell motility during in vitro injury resolution.
Collapse
Affiliation(s)
- K M Providence
- Cell & Molecular Biology Program, Albany Medical College, Albany, NY 12208, USA
| | | | | | | |
Collapse
|
46
|
Abstract
Intimal hyperplasia is the process by which the cell population increases within the innermost layer of the arterial wall, such as occurs physiologically during closure of the ductus arteriosus and during involution of the uterus. It also occurs pathologically in pulmonary hypertension, atherosclerosis, after angioplasty, in transplanted organs, and in vein grafts. The underlying causes of intimal hyperplasia are migration and proliferation of vascular smooth muscle cells provoked by injury, inflammation, and stretch. This review discusses, at a molecular level, both the final common pathways leading to smooth muscle migration and proliferation and their (patho)-physiological triggers. It emphasizes the key roles played by growth factors and extracellular matrix-degrading metalloproteinases, which act in concert to remodel the extracellular matrix and permit cell migration and proliferation.
Collapse
Affiliation(s)
- A C Newby
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| | | |
Collapse
|
47
|
Ahn MY, Zhang ZG, Tsang W, Chopp M. Endogenous plasminogen activator expression after embolic focal cerebral ischemia in mice. Brain Res 1999; 837:169-76. [PMID: 10433999 DOI: 10.1016/s0006-8993(99)01645-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Urokinase-type plasminogen activator (u-PA) and tissue-type plasminogen activator (t-PA) play important roles in fibrinolysis, cell migration, tissue destruction, angiogenesis and tissue remodeling. u-PA and t-PA activity in tissue are tightly regulated by plasminogen activator inhibitor-1 (PAI-1). However, little is known of the activity of endogenous plasminogen activators (PAs) and PAI-1 in ischemic brain. To evaluate whether cerebral ischemic injury induces endogenous PAs and PAI-1, we measured PA activity from brain homogenates, and examined the expression of t-PA mRNA, u-PA mRNA and PAI-1 mRNA from brain homogenates in C57BL/6J mice (n=45) weighing 29-35 g in which the middle cerebral artery (MCA) was occluded by a fibrin-rich clot. Brain homogenates were prepared for direct casein zymography from control non-ischemic mice (n=4) and mice at 2 h (n=5), 4 h (n=5), and 24 h (n=4) after MCA occlusion (MCAO). Also, u-PA and t-PA knockout mice at 4 h (n=2, each) after MCAO were used as a negative control for direct casein zymography. Frozen sections for in situ zymography were obtained from control mice (n=2) and mice at 2 h, 4 h, and 24 h (n=2, per time point) after clot occlusion. Brain homogenates were prepared for reverse transcriptase-polymerase chain reaction (RT-PCR) to examine t-PA mRNA, u-PA mRNA and PAI-1 mRNA expression from control non-ischemic mice (n=4) and mice at 2 h (n=5), 4 h (n=5), and 24 h (n=5) after MCAO. By direct casein zymography, u-PA activity increased at 4 h (P<0.05), and 24 h (P<0.05) after stroke in the ischemic hemisphere compared with the non-ischemic mice. Activity of t-PA in ischemic brain was not significantly different from the control group. As measured by in situ zymography, PA activity, most likely u-PA, was present in the ischemic hemisphere. By RT-PCR, expression of PAI-1 mRNA, but not u-PA mRNA and t-PA mRNA, increased 3-, 15- and 25-folds in the ischemic hemisphere at 2 h, 4 h and 24 h after stroke, respectively, compared with control mice. This study demonstrates that PAI-1 mRNA and u-PA activity increase in mouse brain after stroke.
Collapse
Affiliation(s)
- M Y Ahn
- Department of Neurology, Soonchunhyang University Hospital, Seoul, South Korea
| | | | | | | |
Collapse
|
48
|
Werner F, Razzaq TM, Ellis V. Tissue plasminogen activator binds to human vascular smooth muscle cells by a novel mechanism. Evidence for a reciprocal linkage between inhibition of catalytic activity and cellular binding. J Biol Chem 1999; 274:21555-61. [PMID: 10419460 DOI: 10.1074/jbc.274.31.21555] [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: 11/06/2022] Open
Abstract
Human vascular smooth muscle cells (VSMC) bind tissue plasminogen activator (tPA) specifically, saturably, and with relatively high affinity (K(d) 25 nM), and this binding potentiates the activation of cell-associated plasminogen (Ellis, V., and Whawell, S. A. (1997) Blood 90, 2312-2322). We have observed that this binding can be efficiently competed by DFP-inactivated tPA and S478A-tPA but not by tPA inactivated with H-D-Phe-Pro-Arg-chloromethyl ketone (PPACK). VSMC-bound tPA also exhibited a markedly reduced inhibition by PPACK, displaying biphasic kinetics with second-order rate constants of 7. 5 x 10(3) M(-1) s(-1) and 0.48 x 10(3) M(-1) s(-1), compared with 7. 2 x 10(3) M(-1) s(-1) in the solution phase. By contrast, tPA binding to fibrin was competed equally well by all forms of tPA, and its inhibition was unaltered. These effects were shown to extend to the physiological tPA inhibitor, plasminogen activator inhibitor 1. tPA.plasminogen activator inhibitor 1 complex did not compete tPA binding to VSMC, and the inhibition of bound tPA was reduced by 30-fold. The behavior of the various forms of tPA bound to VSMC correlated with conformational changes in tPA detected by CD spectroscopy. These data suggest that tPA binds to its specific high affinity site on VSMC by a novel mechanism involving the serine protease domain of tPA and distinct from its binding to fibrin. Furthermore, reciprocally linked conformational changes in tPA appear to have functionally significant effects on both the interaction of tPA with its VSMC binding site and the susceptibility of bound tPA to inhibition.
Collapse
Affiliation(s)
- F Werner
- Thrombosis Research Institute, Manresa Rd., London SW3 6LR, United Kingdom
| | | | | |
Collapse
|
49
|
Christ G, Kostner K, Zehetgruber M, Binder BR, Gulba D, Huber K. Plasmin activation system in restenosis: role in pathogenesis and clinical prediction? J Thromb Thrombolysis 1999; 7:277-85. [PMID: 10375389 DOI: 10.1023/a:1008983110941] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
During recent years it has become increasingly recognized that the plasmin activation system is involved in the development of atherosclerosis and restenosis. Responsible pathophysiologic mechanisms, however, remain elusive. This review focuses primarily on the clinicians, point of view, suggesting that increases in plasminogen activator inhibitor type-1 (PAI-1) plasma levels after balloon angioplasty or permanently elevated lipoprotein (a) (Lp(a)) plasma levels might be helpful in the prediction of restenosis after coronary angioplasty. In contrast, tissue-type plasminogen activator (tPA) plasma levels appear unrelated to restenosis, and data regarding a possible role of urokinase-type plasminogen activator (uPA) in circulation are not available at present. Furthermore, a new hypothesis on the pathophysiological role of local PAI-1 overexpression as a beneficial negative feedback mechanism to limit excess cellular proliferation in atherogenesis and restenosis is presented.
Collapse
Affiliation(s)
- G Christ
- Department of Cardiology, University of Vienna, Austria
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
The epidermis is a self-renewing tissue that must maintain a basal proliferative rate as well as respond to various perturbing stimuli. Regulation of keratinocyte proliferation involves diverse molecules, including growth factors, ions, and hormones. We recently proposed that a proteinase, urokinase-type plasminogen activator (uPA) may be added to this list, based on correlative evidence linking expression of uPA and murine epidermal hyperproliferation. Here we report that, during the first 3 d of life, the epidermis from mice that bear a targeted deletion of the uPA gene has a significantly lower proliferative rate than the epidermis from wild-type mice. In contrast, proliferation in the matrix keratinocytes of the hair follicles is not decreased in neonatal uPA-/- mice. Vertical migration of keratinocytes during terminal differentiation was not affected. We therefore conclude that lack of uPA is associated with a decrease in epidermal proliferation.
Collapse
Affiliation(s)
- P J Jensen
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia 19104-6142, USA
| | | |
Collapse
|