1
|
Lipoprotein(a) in Atherosclerotic Diseases: From Pathophysiology to Diagnosis and Treatment. Molecules 2023; 28:molecules28030969. [PMID: 36770634 PMCID: PMC9918959 DOI: 10.3390/molecules28030969] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is a low-density lipoprotein (LDL) cholesterol-like particle bound to apolipoprotein(a). Increased Lp(a) levels are an independent, heritable causal risk factor for atherosclerotic cardiovascular disease (ASCVD) as they are largely determined by variations in the Lp(a) gene (LPA) locus encoding apo(a). Lp(a) is the preferential lipoprotein carrier for oxidized phospholipids (OxPL), and its role adversely affects vascular inflammation, atherosclerotic lesions, endothelial function and thrombogenicity, which pathophysiologically leads to cardiovascular (CV) events. Despite this crucial role of Lp(a), its measurement lacks a globally unified method, and, between different laboratories, results need standardization. Standard antilipidemic therapies, such as statins, fibrates and ezetimibe, have a mediocre effect on Lp(a) levels, although it is not yet clear whether such treatments can affect CV events and prognosis. This narrative review aims to summarize knowledge regarding the mechanisms mediating the effect of Lp(a) on inflammation, atherosclerosis and thrombosis and discuss current diagnostic and therapeutic potentials.
Collapse
|
2
|
Kroone C, Vos M, Rademakers T, Kuijpers M, Hoogenboezem M, van Buul J, Heemskerk JWM, Ruf W, van Hylckama Vlieg A, Versteeg HH, Goumans MJ, de Vries CJM, Kurakula K. LIM-only protein FHL2 attenuates vascular tissue factor activity, inhibits thrombus formation in mice and FHL2 genetic variation associates with human venous thrombosis. Haematologica 2019; 105:1677-1685. [PMID: 31467128 PMCID: PMC7271603 DOI: 10.3324/haematol.2018.203026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/26/2019] [Indexed: 12/21/2022] Open
Abstract
Bleeding disorders and thrombotic complications are major causes of morbidity and mortality with many cases being unexplained. Thrombus formation involves aberrant expression and activation of tissue factor (TF) in vascular endothelial and smooth muscle cells. Here, we sought to identify factors that modulate TF gene expression and activity in these vascular cells. The LIM-only protein FHL2 is a scaffolding protein that modulates signal transduction pathways with crucial functions in endothelial and smooth muscle cells. However, the role of FHL2 in TF regulation and thrombosis remains unexplored. Using a murine model of venous thrombosis in mesenteric vessels, we demonstrated that FHL2 deficiency results in exacerbated thrombus formation. Gain- and loss-of-function experiments revealed that FHL2 represses TF expression in endothelial and smooth muscle cells through inhibition of the transcription factors nuclear factor κB and activating protein-1. Furthermore, we observed that FHL2 interacts with the cytoplasmic tail of TF. In line with our in vivo observations, FHL2 decreases TF activity in endothelial and smooth muscle cells whereas FHL2 knockdown or deficiency results in enhanced TF activity. Finally, the FHL2 single nucleotide polymorphism rs4851770 was associated with the risk of venous thrombosis in a large population of venous thrombosis cases and control subjects from 12 studies (INVENT consortium). Altogether, our results highlight functional involvement of FHL2 in TF-mediated coagulation and identify FHL2 as a novel gene associated with venous thrombosis in humans.
Collapse
Affiliation(s)
- Chantal Kroone
- The Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center (UMC), Leiden, the Netherlands
| | - Mariska Vos
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Timo Rademakers
- Department of Molecular Cell Biology, Sanquin Research, Amsterdam, the Netherlands
| | - Marijke Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC, Maastricht, The Netherlands
| | - Mark Hoogenboezem
- Department of Molecular Cell Biology, Sanquin Research, Amsterdam, the Netherlands
| | - Jaap van Buul
- Department of Molecular Cell Biology, Sanquin Research, Amsterdam, the Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UMC, Maastricht, The Netherlands
| | - Wolfram Ruf
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.,Center for Thrombosis and Hemostasis Mainz, Germany
| | | | - Henri H Versteeg
- The Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center (UMC), Leiden, the Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Carlie J M de Vries
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kondababu Kurakula
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands .,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
3
|
Yeh YH, Chang SH, Chen SY, Wen CJ, Wei FC, Tang R, Achilefu S, Wun TC, Chen WJ. Bolus injections of novel thrombogenic site-targeted fusion proteins comprising annexin-V and Kunitz protease inhibitors attenuate intimal hyperplasia after balloon angioplasty. Int J Cardiol 2017; 240:339-346. [PMID: 28433556 DOI: 10.1016/j.ijcard.2017.03.150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 02/26/2017] [Accepted: 03/27/2017] [Indexed: 11/13/2022]
Abstract
BACKGROUND Systemic administrations of conventional antithrombotics reduce neointima formation after angioplasty in experimental animals. However, clinical translation of these results has not been successful due to high risk for bleeding. OBJECTIVES We sought to determine whether novel annexin-V (ANV)-Kunitz protease inhibitor fusion proteins, TAP-ANV and ANV-6L15, can specifically target to vascular injury site and limit neointima formation without inducing systemic hypo-coagulation in a rat carotid artery balloon angioplasty injury model. METHODS Near infrared imaging was carried out after balloon-injury and injection of fluorescent ANV or ANV-6L15 to examine their bio-distributions. For peri-procedure treatment, TAP-ANV or ANV-6L15 was administered as i.v. boluses 3 times: 30-minutes before balloon-injury, immediate after procedure, and 120-minutes post-balloon-injury. For extended treatment, additional i.v. bolus injection was given on day-2, day-3 and every other day thereafter. Carotid arteries were collected on day-7 and day-14 for analysis. Blood was collected for measurement of clotting parameters. RESULTS Near infrared imaging and immunochemistry showed that fluorescent ANV and ANV-6L15 specifically localized to injured carotid artery and significant amount of ANV-6L15 remained bound to the injured artery after 24-h. Peri-procedure injections of TAP-ANV or ANV-6L15 resulted in decrease of intima/media ratio by 56%. Extended injections of both yielded similar results. Both decreased the expression of PCNA on day-7 and increased the expression calponin on day-14 in the intima post-balloon-injury. CONCLUSIONS TAP-ANV and ANV-6L15 can specifically localize to balloon injured carotid arteries after i.v. bolus injections, resulting in substantial attenuation of intimal hyperplasia without inducing a state of systemic hypo-coagulation.
Collapse
Affiliation(s)
- Yung-Hsin Yeh
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan.
| | - Shang-Hung Chang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Shin-Yu Chen
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Chih-Jen Wen
- College of Medicine, Chang-Gung University, Center for Vascularized Composite Allotransplantation, Chang-Gung Medical Foundation, Department of Plastic and Reconstructive Surgery and Center for Vascularized Composite Allotransplantation, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Fu-Chan Wei
- College of Medicine, Chang-Gung University, Center for Vascularized Composite Allotransplantation, Chang-Gung Medical Foundation, Department of Plastic and Reconstructive Surgery and Center for Vascularized Composite Allotransplantation, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Rui Tang
- Department of Radiology, Washington University Medical School, St Louis, MO 63110, USA
| | - Sam Achilefu
- Department of Radiology, Washington University Medical School, St Louis, MO 63110, USA
| | - Tze-Chein Wun
- EVAS Therapeutics, LLC, 613 Huntley Heights Drive, Ballwin, MO 63021, USA
| | - Wei-Jan Chen
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
4
|
Assessment of Novel Anti-thrombotic Fusion Proteins for Inhibition of Stenosis in a Porcine Model of Arteriovenous Graft. PLoS One 2015; 10:e0137381. [PMID: 26360605 PMCID: PMC4567316 DOI: 10.1371/journal.pone.0137381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 08/16/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hemodialysis arteriovenous synthetic grafts (AVG) provide high volumetric blood flow rates shortly after surgical placement. However, stenosis often develops at the vein-graft anastomosis contributing to thrombosis and early graft failure. Two novel fusion proteins, ANV-6L15 and TAP-ANV, inhibit the tissue factor/factor VIIa coagulation complex and the factor Xa/factor Va complex, respectively. Each inhibitor domain is fused to an annexin V domain that targets the inhibitor activity to sites of vascular injury to locally inhibit thrombosis. This study's objective was to determine if these antithrombotic proteins are safe and effective in inhibiting AVG stenosis. METHODS A bolus of either TAP-ANV or ANV-6L15 fusion protein was administered intravenously immediately prior to surgical placement of a synthetic graft between the external jugular vein and common carotid artery in a porcine model. At surgery, the vein and artery were irrigated with the anti-thrombotic fusion protein. Control animals received intravenous heparin. At 4 weeks, MRI was performed to evaluate graft patency, the pigs were then euthanized and grafts and attached vessels were explanted for histomorphometric assessment of neointimal hyperplasia at the vein-graft anastomosis. Blood was collected at surgery, immediately after surgery and at euthanasia for serum metabolic panels and coagulation chemistries. RESULTS No acute thrombosis occurred in the control group or in either experimental group. No abnormal serum chemistries, activated clotting times or PT, PTT values were observed after treatment in experimental or control animals. However, at the vein-graft anastomosis, there was no difference between the control and experimental groups in cross-sectional lumen areas, as measured on MRI, and no difference in hyperplasia areas as determined by histomorphometry. These results suggest that local irrigation of TAP-ANV or ANV-6L15 intra-operatively was as effective in inhibiting acute graft thrombosis as intravenous administration of heparin, but failed to inhibit hyperplasia development and stenosis in AVG.
Collapse
|
5
|
Winckers K, ten Cate H, Hackeng TM. The role of tissue factor pathway inhibitor in atherosclerosis and arterial thrombosis. Blood Rev 2013; 27:119-32. [PMID: 23631910 DOI: 10.1016/j.blre.2013.03.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tissue factor pathway inhibitor (TFPI) is the main inhibitor of tissue factor (TF)-mediated coagulation. In atherosclerotic plaques TFPI co-localizes with TF, where it is believed to play an important role in attenuating TF activity. Findings in animal models such as TFPI knockout models and gene transfer models are consistent on the role of TFPI in arterial thrombosis as they reveal an active role for TFPI in attenuating arterial thrombus formation. In addition, ample experimental evidence exists indicating that TFPI has inhibitory effects on both smooth muscle cell migration and proliferation, both which are recognized as important pathological features in atherosclerosis development. Nonetheless, the clinical relevance of these antithrombotic and atheroprotective effects remains unclear. Paradoxically, the majority of clinical studies find increased instead of decreased TFPI antigen and activity levels in atherothrombotic disease, particularly in atherosclerosis and coronary artery disease (CAD). Increased TFPI levels in cardiovascular disease might result from complex interactions with established cardiovascular risk factors, such as hypercholesterolemia, diabetes and smoking. Moreover, it is postulated that increased TFPI levels reflect either the amount of endothelial perturbation and platelet activation, or a compensatory mechanism for the increased procoagulant state observed in cardiovascular disease. In all, the prognostic value of plasma TFPI in cardiovascular disease remains to be established. The current review focuses on TFPI in clinical studies of asymptomatic and symptomatic atherosclerosis, coronary artery disease and ischemic stroke, and discusses potential atheroprotective actions of TFPI.
Collapse
Affiliation(s)
- Kristien Winckers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, MUMC, Maastricht, The Netherlands
| | | | | |
Collapse
|
6
|
Petrillo G, Cirillo P, D'Ascoli GL, Maresca F, Ziviello F, Chiariello M. Tissue Factor/Factor FVII Complex Inhibitors in Cardiovascular Disease. Are Things Going Well? Curr Cardiol Rev 2011; 6:325-32. [PMID: 22043208 PMCID: PMC3083813 DOI: 10.2174/157340310793566190] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 06/17/2010] [Accepted: 06/22/2010] [Indexed: 11/22/2022] Open
Abstract
Blood coagulation is a complex biological mechanism aimed to avoid bleeding in which a highly regulated and coordinated interplay of specific proteins and cellular components respond quickly to a vascular injury. However, when this mechanisms occurs in the coronary circulation, it has not a “protective” effect, but rather, it plays a pivotal role in determining acute coronary syndromes. Coagulation recognizes Tissue Factor (TF), the main physiological initiator of the extrinsic coagulation pathway, as its starter. Since TF:VIIa complex is the critical point of the blood coagulation cascade, it is a pharmacological attractive issue for the development of agents with anti thrombotic properties that can exert their activity by inhibiting complex formation and/or its catalytic activity. In fact, it is intuitive that an antithrombotic agent able to inhibit this initial step of the coagulation pathway has several theoretical, extremely important, advantages if compared with drugs active downstream the coagulation pathway, such as FXa or thrombin. The present report gives a brief overview of TF pathophysiology, highlighting the most recent advances in the field of inhibitors of the complex TF/VIIa potentially useful in cardiovascular disease.
Collapse
Affiliation(s)
- Gianluca Petrillo
- Department of Internal Medicine, Cardiovascular and Immunological Sciences (Division of Cardiology) University of Naples "Federico II", Italy
| | | | | | | | | | | |
Collapse
|
7
|
Affiliation(s)
- Julian Ilcheff Borissoff
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute of Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | |
Collapse
|
8
|
Zhou J, Wang Y, Xiong Y, Wang H, Feng Y, Chen J. Delivery of TFPI-2 using ultrasound with a microbubble agent (SonoVue) inhibits intimal hyperplasia after balloon injury in a rabbit carotid artery model. ULTRASOUND IN MEDICINE & BIOLOGY 2010; 36:1876-1883. [PMID: 20888684 DOI: 10.1016/j.ultrasmedbio.2010.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 08/13/2010] [Accepted: 08/21/2010] [Indexed: 05/29/2023]
Abstract
Here we report a new, simple and efficient method by using ultrasound and a microbubble agent (SonoVue) for delivering a gene to balloon-injured carotid arteries for restenosis prophylaxis. The tissue factor pathway inhibitor-2 (TFPI-2) has been shown to inhibit the postinjury intimae hyperplasia in atherosclerotic vessels. New Zealand white rabbits were divided into 4 groups with 14 in each, a treatment control for balloon injury, a gene vehicle control, a gene delivery of TFPI-2 without using ultrasound and a gene delivery of TFPI-2 using ultrasound. After four weeks, the injured artery neointimal proliferation was significantly lower in the TFPI-2 group with ultrasound than the control groups (p < 0.01) according to the measurement of the mean luminal diameters by B-mode ultrasonography. The ratio of intimal/media area and the stenosis rate in the gene delivery facilitated by ultrasound were significantly lower than those of the nonultrasound gene delivering method (p < 0.01).
Collapse
Affiliation(s)
- Jie Zhou
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
9
|
He X, Han B, Mura M, Li L, Cypel M, Soderman A, Picha K, Yang J, Liu M. Anti-human tissue factor antibody ameliorated intestinal ischemia reperfusion-induced acute lung injury in human tissue factor knock-in mice. PLoS One 2008; 3:e1527. [PMID: 18231608 PMCID: PMC2211395 DOI: 10.1371/journal.pone.0001527] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Accepted: 12/27/2007] [Indexed: 01/11/2023] Open
Abstract
Background Interaction between the coagulation and inflammation systems plays an important role in the development of acute respiratory distress syndrome (ARDS). Anti-coagulation is an attractive option for ARDS treatment, and this has promoted development of new antibodies. However, preclinical trials for these antibodies are often limited by the high cost and availability of non-human primates. In the present study, we developed a novel alternative method to test the role of a humanized anti-tissue factor mAb in acute lung injury with transgenic mice. Methodology/Principal Findings Human tissue factor knock-in (hTF-KI) transgenic mice and a novel humanized anti-human tissue factor mAb (anti-hTF mAb, CNTO859) were developed. The hTF-KI mice showed a normal and functional expression of hTF. The anti-hTF mAb specifically blocked the pro-coagulation activity of brain extracts from the hTF-KI mice and human, but not from wild type mice. An extrapulmonary ARDS model was used by intestinal ischemia-reperfusion. Significant lung tissue damage in hTF-KI mice was observed after 2 h reperfusion. Administration of CNTO859 (5 mg/kg, i.v.) attenuated the severity of lung tissue injury, decreased the total cell counts and protein concentration in bronchoalveolar lavage fluid, and reduced Evans blue leakage. In addition, the treatment significantly reduced alveolar fibrin deposition, and decreased tissue factor and plasminogen activator inhibitor-1 activity in the serum. This treatment also down-regulated cytokine expression and reduced cell death in the lung. Conclusions This novel anti-hTF antibody showed beneficial effects on intestinal ischemia-reperfusion induced acute lung injury, which merits further investigation for clinical usage. In addition, the use of knock-in transgenic mice to test the efficacy of antibodies against human-specific proteins is a novel strategy for preclinical studies.
Collapse
Affiliation(s)
- Xiaolin He
- Latner Thoracic Surgery Research Laboratories, Department of Surgery, University Health Network Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Bing Han
- Latner Thoracic Surgery Research Laboratories, Department of Surgery, University Health Network Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Marco Mura
- Latner Thoracic Surgery Research Laboratories, Department of Surgery, University Health Network Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Li Li
- Therakos, Exton, Pennsylvania, United State of America
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, Department of Surgery, University Health Network Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Avery Soderman
- Centocor Inc., Malvern, Pennsylvania, United States of America
| | - Kristen Picha
- Centocor Inc., Malvern, Pennsylvania, United States of America
| | - Jing Yang
- Centocor Inc., Malvern, Pennsylvania, United States of America
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Department of Surgery, University Health Network Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
10
|
Niiro M, Nagayama T, Yunoue S, Obara S, Hirano H. Changes in tissue factor and the effects of tissue factor pathway inhibitor on transient focal cerebral ischemia in rats. Thromb Res 2007; 122:247-55. [PMID: 18067952 DOI: 10.1016/j.thromres.2007.10.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 10/01/2007] [Accepted: 10/03/2007] [Indexed: 11/29/2022]
Abstract
INTRODUCTION To determine the contribution of tissue factor (TF) to focal cerebral ischemia/reperfusion injury, we investigated the changes in TF in rat brains with transient focal cerebral ischemia and also assessed the effect of TF pathway inhibitor (TFPI). MATERIALS AND METHODS Spontaneous hypertensive rats were subjected to 90-min of middle cerebral artery occlusion (MCAO) and then were reperfused for up to 24 h. Immediately after MCAO, recombinant human TFPI (rhTFPI) (50 or 20 microg/kg/min) was administered by means of a continuous intravenous injection for 4.5 h. RESULTS AND CONCLUSIONS TF immunoreactivity decreased or scattered in the ischemic area after reperfusion, however, an increased TF expression was observed in the microvasculature with the surrounding brain parenchyma and it peaked at 3 to 6 h, which coincided with the start of fibrin formation. On the other hand, total TF protein in ischemic area continued to exist and did not remarkably change until 24 h after reperfusion. At 24 h after reperfusion, the total infarct volume in the group treated with 50 microg/kg/min rhTFPI was significantly smaller than that in the controls (saline). Western blotting and immunohistochemical studies showed that rhTFPI treatment resulted in a decrease of fibrin in the ischemic brains and microvasculature. TF-mediated microvascular thrombosis is thus considered to contribute to focal cerebral ischemia/reperfusion injury. The continuous infusion of rhTFPI until a peak of TF-mediated microvascular thrombosis therefore attenuates the infarct volume by reducing fibrin deposition in the cerebral microcirculation.
Collapse
Affiliation(s)
- Masaki Niiro
- Department of Neurosurgery, Graduate School of Medicine and Dental Sciences, University of Kagoshima, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan.
| | | | | | | | | |
Collapse
|
11
|
Wallitt EJW, Jevon M, Hornick PI. Therapeutics of vein graft intimal hyperplasia: 100 years on. Ann Thorac Surg 2007; 84:317-23. [PMID: 17588453 DOI: 10.1016/j.athoracsur.2007.02.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Revised: 02/13/2007] [Accepted: 02/15/2007] [Indexed: 11/26/2022]
Abstract
Intimal hyperplasia is central to the pathology of vein graft re-stenosis, and despite considerable advances in our understanding of vascular biology since it was first described 100 years ago, it is still a significant clinical problem. Recent decades have seen the development of many new therapeutic agents aimed at treating this condition, but the successes of laboratory studies have not been replicated in the clinic yet. This review discusses these therapeutic agents, how their modes of action relate to the pathogenesis of vein graft intimal hyperplasia, and considerations of ways in which such therapy may be improved in the future.
Collapse
Affiliation(s)
- Edward J W Wallitt
- Department of Cardiothoracic Surgery, Imperial College, London, United Kingdom
| | | | | |
Collapse
|
12
|
Wickline SA, Neubauer AM, Winter PM, Caruthers SD, Lanza GM. Molecular imaging and therapy of atherosclerosis with targeted nanoparticles. J Magn Reson Imaging 2007; 25:667-80. [PMID: 17347992 DOI: 10.1002/jmri.20866] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Advances in bionanotechnology are poised to impact the field of cardiovascular diagnosis and therapy for decades to come. This review seeks to illustrate selected examples of newly developed diagnostic and therapeutic nanosystems that have been evaluated in experimental atherosclerosis, thrombosis, and vascular biology. We review a variety of nanotechnologies that are capable of detecting early cardiovascular pathology, as well as associated imaging approaches and conjunctive strategies for site-targeted treatment with nanoparticle delivery systems.
Collapse
Affiliation(s)
- Samuel A Wickline
- Department of Medicine, Washington University, St. Louis, Missouri 63110, USA.
| | | | | | | | | |
Collapse
|
13
|
Völzke H, Rettig R. Present status of outcome prediction of invasive coronary treatment by using genetic markers. Hum Mutat 2006; 27:307-22. [PMID: 16511827 DOI: 10.1002/humu.20305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A growing number of studies suggest that the outcome after invasive coronary treatment may be in part genetically determined. Here, we review the present status of outcome prediction of invasive coronary treatments by using genetic markers. Although some studies found an association between one or another genetic marker with one or another clinical endpoint, many other studies found no such relations; to date, none of the genetic markers that have been investigated in association studies are used in routine clinical practice to prospectively assess the prognosis following invasive coronary treatment or to decide upon therapeutic strategies. Many associations between genetic markers and certain clinical endpoints were initially reported in small studies but could not be confirmed in larger ones. Some of these discrepancies may be explained by publication bias. Some genetic variants may have true effects on clinical endpoints, which, albeit biologically interesting, do not bear much clinical relevance. On the other hand, many-if not most-studies that have been published to date are more or less grossly underpowered and very rarely report on the results of an a priori power analysis. Thus, there is still a need for further high-quality studies designed to investigate the specific contribution of genetic factors to the outcome after invasive coronary interventions.
Collapse
Affiliation(s)
- Henry Völzke
- Institute of Epidemiology and Social Medicine, Ernst Moritz Arndt University, Greifswald, Germany.
| | | |
Collapse
|
14
|
Bogdanov VY, Kirk RI, Miller C, Hathcock JJ, Vele S, Gazdoiu M, Nemerson Y, Taubman MB. Identification and characterization of murine alternatively spliced tissue factor. J Thromb Haemost 2006; 4:158-67. [PMID: 16409465 DOI: 10.1111/j.1538-7836.2005.01680.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tissue factor (TF) is a transmembrane glycoprotein that initiates coagulation and plays a critical role in regulating hemostasis and thrombosis. We have recently reported a naturally occurring, soluble form of human tissue factor (asTF) generated by alternative splicing. This splice variant has a novel C-terminus with no homology to that of the full-length TF (flTF), lacks a transmembrane domain, and is active in the presence of phospholipids. Mouse models offer unique opportunities to examine the relative importance of flTF and asTF in mediating thrombosis, the response to arterial injury, and ischemic damage. To that end, we have identified and characterized murine asTF (masTF). Like the human splice variant, masTF lacks a transmembrane domain and has a unique C-terminus. We have generated antibodies specific to masTF and murine flTF (mflTF) to examine the expression of both forms of TF. masTF antigen is widely and abundantly expressed, with a pattern similar to that of mflTF, in adult tissues, in experimentally induced thrombi, and during development. These studies demonstrate that masTF contributes to the pool of total TF and may thus play an important role in mediating TF-dependent processes.
Collapse
Affiliation(s)
- V Y Bogdanov
- Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Chen HH, Vicente CP, He L, Tollefsen DM, Wun TC. Fusion proteins comprising annexin V and Kunitz protease inhibitors are highly potent thrombogenic site-directed anticoagulants. Blood 2005; 105:3902-9. [PMID: 15677561 PMCID: PMC1895074 DOI: 10.1182/blood-2004-11-4435] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2004] [Accepted: 01/12/2005] [Indexed: 11/20/2022] Open
Abstract
The anionic phospholipid, phosphatidyl-L-serine (PS), is sequestered in the inner layer of the plasma membrane in normal cells. Upon injury, activation, and apoptosis, PS becomes exposed on the surfaces of cells and sheds microparticles, which are procoagulant. Coagulation is initiated by formation of a tissue factor/factor VIIa complex on PS-exposed membranes and propagated through the assembly of intrinsic tenase (factor VIIIa/factor IXa), prothrombinase (factor Va/factor Xa), and factor XIa complexes on PS-exposed activated platelets. We constructed a novel series of recombinant anticoagulant fusion proteins by linking annexin V (ANV), a PS-binding protein, to the Kunitz-type protease inhibitor (KPI) domain of tick anticoagulant protein, an aprotinin mutant (6L15), amyloid beta-protein precursor, or tissue factor pathway inhibitor. The resulting ANV-KPI fusion proteins were 6- to 86-fold more active than recombinant tissue factor pathway inhibitor and tick anticoagulant protein in an in vitro tissue factor-initiated clotting assay. The in vivo antithrombotic activities of the most active constructs were 3- to 10-fold higher than that of ANV in a mouse arterial thrombosis model. ANV-KPI fusion proteins represent a new class of anticoagulants that specifically target the anionic membrane-associated coagulation enzyme complexes present at sites of thrombogenesis and are potentially useful as antithrombotic agents.
Collapse
Affiliation(s)
- Hsiu-Hui Chen
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | | |
Collapse
|
16
|
Kaiser B, Hoppensteadt DA, Fareed J. Tissue factor pathway inhibitor for cardiovascular disorders. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728214.5.1.73] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
17
|
Affiliation(s)
- Robina Matyal
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | |
Collapse
|
18
|
Segev A, Hegele RA, Lau HK, Sparkes JD, Teitel JM, Chisholm RJ, Strauss BH. Thr325Ile polymorphism of the TAFI gene is related to TAFI antigen plasma levels and angiographic restenosis after percutaneous coronary interventions. Thromb Res 2004; 114:137-41. [PMID: 15306156 DOI: 10.1016/j.thromres.2004.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2003] [Revised: 06/01/2004] [Accepted: 06/01/2004] [Indexed: 11/16/2022]
Abstract
BACKGROUND The fibrinolytic system is closely related to several processes that are involved in restenosis. We have previously shown that high pre-procedural plasma levels of thrombin-activatable fibrinolysis inhibitor (TAFI) antigen predicted angiographic restenosis. The aims of this study were to evaluate the relationship between Thr325Ile polymorphisms, plasma levels of TAFI antigen, and late angiographic restenosis after percutaneous coronary intervention (PCI). METHODS We prospectively studied 159 patients with stable angina who underwent successful elective angioplasty or stenting of de novo native coronary artery lesions. Blood samples were drawn before the procedure. TAFI antigen levels were measured, as well as the presence of TAFI Ile325Thr genetic variation. Follow-up coronary angiography was performed in 90% of patients. RESULTS The genotypes based on Ile325Thr substitution had significantly different TAFI antigen levels: genotype C/C>C/T>T/T (111+/-29%, 87+/-24%, and 59+/-22%, respectively, p<0.006). T/T genotype was associated with lower rates of restenosis compared to C/T and C/C genotypes (25% versus 37% and 33%, respectively, p<0.05). CONCLUSIONS These data suggest that plasma TAFI antigen levels are genetically controlled. The T/T Thr325Ile polymorphism of the TAFI gene is associated with lower plasma levels of TAFI antigen and lower restenosis rates after PCI.
Collapse
Affiliation(s)
- Amit Segev
- The Roy and Ann Foss Cardiovascular Research Program, Division of Cardiology, Terrence Donnelly Heart Center, St. Michael's Hospital, 30 Bond Street, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | | | | | | | | | | | | |
Collapse
|
19
|
Koh KK, Chung WJ, Ahn JY, Han SH, Kang WC, Seo YH, Ahn TH, Choi IS, Shin EK. Angiotensin II type 1 receptor blockers reduce tissue factor activity and plasminogen activator inhibitor type-1 antigen in hypertensive patients: a randomized, double-blind, placebo-controlled study. Atherosclerosis 2004; 177:155-60. [PMID: 15488878 DOI: 10.1016/j.atherosclerosis.2004.07.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2004] [Accepted: 07/02/2004] [Indexed: 10/26/2022]
Abstract
Angiotensin II stimulates the expression of tissue factor (TF) and plasminogen activator inhibitor type-1 (PAI-1), and AT1 receptor blockade (ARB) reduces PAI-1 and TF activities in experimental studies. We investigated the effects of ARBs on TF activity, tissue plasminogen activator (tPA), PAI-1 antigen levels, plasma renin activity (PRA) and aldosterone levels in hypertensive patients. Placebo, losartan 100mg, irbesartan 300 mg, and candesartan 16 mg daily were administered to 122 patients for 2 months. Compared with placebo, ARBs significantly reduced TF activity (P <0.001 by ANOVA), and candesartan was the most potent. Compared with placebo or losartan, irbesartan and candesartan significantly lowered plasma levels of PAI-1 antigen (P <0.001 by ANOVA) with no differences between the two. Compared with placebo, all ARBs lowered plasma levels of aldosterone (P=0.012 by ANOVA) and increased PRA (P=0.005 by ANOVA). There were significant correlations between the degree of change in TF activity and PAI-1 antigen levels (r=0.458, P <0.0001) and between the change in TF activity and PRA (r=-0.296, P=0.006), but not with the magnitude of reduction in blood pressure following ARB therapy. ARBs significantly reduced TF activity, PAI-1 antigen levels, and aldosterone levels in hypertensive patients. The clinical significance of the varying potency of some ARBs needs to be further investigated.
Collapse
Affiliation(s)
- Kwang Kon Koh
- Vascular Medicine and Atherosclerosis Unit, Cardiology, Gil Heart Center, Gachon Medical School, 1198 Kuwol-dong, Namdong-gu, Incheon 405-760, Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lee MS, David EM, Makkar RR, Wilentz JR. Molecular and cellular basis of restenosis after percutaneous coronary intervention: the intertwining roles of platelets, leukocytes, and the coagulation-fibrinolysis system. J Pathol 2004; 203:861-70. [PMID: 15258987 DOI: 10.1002/path.1598] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The major limitation of percutaneous coronary intervention (PCI) is restenosis. Restenosis is considered to be an overreaction of the natural healing process after traumatic balloon dilatation. An elaborate web of cellular and molecular responses, including the interaction of platelets, leukocytes, and the coagulation-fibrinolysis system, as well as the secretion of various growth factors and pro-inflammatory cytokines, contributes to neointimal hyperplasia and the development of restenosis. Moreover, platelet and neutrophil activation after stenting appears to be different from that after balloon angioplasty alone. Pharmacotherapy targeting the cell-to-cell interaction between platelets and neutrophils may potentially offer an effective treatment strategy against restenosis after PCI.
Collapse
Affiliation(s)
- Michael S Lee
- Cedars-Sinai Medical Center, UCLA School of Medicine, Cardiovascular Intervention Center, Los Angeles, California, USA.
| | | | | | | |
Collapse
|
21
|
Morawski AM, Winter PM, Crowder KC, Caruthers SD, Fuhrhop RW, Scott MJ, Robertson JD, Abendschein DR, Lanza GM, Wickline SA. Targeted nanoparticles for quantitative imaging of sparse molecular epitopes with MRI. Magn Reson Med 2004; 51:480-6. [PMID: 15004788 DOI: 10.1002/mrm.20010] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Before molecular imaging with MRI can be applied clinically, certain problems, such as the potential sparseness of molecular epitopes on targeted cell surfaces, and the relative weakness of conventional targeted MR contrast agents, must be overcome. Accordingly, the conditions for diagnostic conspicuity that apply to any paramagnetic MRI contrast agent with known intrinsic relaxivity were examined in this study. A highly potent paramagnetic liquid perfluorocarbon nanoparticle contrast agent ( approximately 250 nm diameter, >90,000 Gd3+/particle) was imaged at 1.5 T and used to successfully predict a range of sparse concentrations in experimental phantoms with the use of standard MR signal models. Additionally, we cultured and targeted the smooth muscle cell (SMC) monolayers that express "tissue factor," a glycoprotein of crucial significance to hemostasis and response to vascular injury, by conjugating an anti-tissue factor antibody fragment to the nanoparticles to effect specific binding. Quantification of the signal from cell monolayers imaged at 1.5 T demonstrated, as predicted via modeling, that only picomolar concentrations of paramagnetic perfluorocarbon nanoparticles were required for the detection and quantification of tissue factor at clinical field strengths. Thus, for targeted paramagnetic agents carrying high payloads of gadolinium, it is possible to quantify molecular epitopes present in picomolar concentrations in single cells with routine MRI.
Collapse
Affiliation(s)
- Anne M Morawski
- Cardiovascular MR Laboratories, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Targeted contrast agents are expanding the detectability and diagnosis of pathology from a strict anatomic to biochemical basis. Moreover, these new agents, in their various forms, offer the potential for site-specific drug and gene delivery, i.e., the "magic bullet" first postulated by Paul Erhlich 100 years ago. The ability to direct drugs to the molecular signatures of disease, to confirm noninvasively their presence at the site-of-interest, and to quantify the adequacy of local drug concentration at the time of treatment, ie, rational targeted drug delivery, offers exciting new clinical paradigms in the near future.
Collapse
|
23
|
Kim WJH, Chereshnev I, Gazdoiu M, Fallon JT, Rollins BJ, Taubman MB. MCP-1 deficiency is associated with reduced intimal hyperplasia after arterial injury. Biochem Biophys Res Commun 2003; 310:936-42. [PMID: 14550294 DOI: 10.1016/j.bbrc.2003.09.088] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monocyte chemoattractant protein (MCP)-1 is abundant in smooth muscle cells (SMC) and macrophages of atherosclerotic plaques and in the injured arterial wall. MCP-1 and its receptor, CCR2, are important mediators of macrophage accumulation and atherosclerotic plaque progression. We have recently reported that CCR2(-/-) mice have a approximately 60% decrease in intimal hyperplasia and medial DNA synthesis in response to femoral arterial injury. We have now examined the response to femoral arterial injury in MCP-1(-/-) mice. MCP-1 deficiency was associated with a approximately 30% reduction in intimal hyperplasia at 4 weeks and was not associated with diminished medial DNA synthesis. Despite inducing tissue factor in SMC culture, MCP-1 deficiency was not associated with a decrease in neointimal tissue factor after injury. These data suggest that MCP-1 and CCR2 deficiencies have distinct effects on arterial injury. The effects of MCP-1 on intimal hyperplasia may be mediated largely through SMC migration.
Collapse
Affiliation(s)
- William J H Kim
- The Zena and Michael A Wiener Cardiovascular Institute, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
24
|
Jeong WK, Jeong MH, Kim KH, Lee SR, Park OY, Yum JH, Kim JH, Kim W, Rhew JY, Ahn YK, Cho JG, Ahn BH, Suh SP, Park JC, Kim SH, Kang JC. An elevated value of C-reactive protein is the only predictive factor of restenosis after percutaneous coronary intervention. Korean J Intern Med 2003; 18:154-60. [PMID: 14619384 PMCID: PMC4531625 DOI: 10.3904/kjim.2003.18.3.154] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The current techniques for percutaneous coronary interventions (PCI) remain limited by restenosis. Recent studies have provided evidence of inflammation playing a role in the pathogenesis of cardiovascular disease. METHODS Whether inflammatory markers are predictors of subsequent restenosis were prospectively tested in 272 consecutive patients with angiographically proven coronary artery disease. Patients having undergone PCI at Chonnam National University Hospital, between Sept. 1999 and Mar. 2001, were divided into two groups according to the occurrence of restenosis on a follow-up coronary angiogram: Group I were patients with restenosis (n = 99, 59.5 +/- 10.8 years, M:F = 77:22) and Group II were those without restenosis (n = 173, 58.8 +/- 10.2 years, M:F = 131:42). The IgG seropositivity, cytomegalovirus (CMV) titers, C. pneumoniae. H. pylori and levels of C-reactive protein (CRP) were compared between the two groups. RESULTS There were no statistical differences in the seropositivity of the CMV IgG C. pneumoniae IgG and H. pylori IgG between the two groups (Groups I vs. II: 100 vs. 100%, 24.7 vs. 25.7% and 62.2 vs. 63.7%, respectively). Of the angiographic parameters, a low Thrombolysis in Myocardial Infarction (TIMI) flow (TIMI 0 or 1) was more common in Group I than Group II (p = 0.038). The patients with an elevated CRP (> 0.5 mg/dL) were more common in Group I than Group II (57.6 vs. 36.4%, p = 0.001), with the CRP values being higher in Group I than Group II (3.3 +/- 5.8 vs. 1.3 +/- 2.6 mg/dL, p = 0.001). According to a multiple logistic regression analysis, the CRP was the only predictor of restenosis, with an odds ratio of 2.1169 (95% C.I. 1.2062-3.7154, p = 0.009). CONCLUSION The CRP value is the most important predictor of restenosis after PCI.
Collapse
Affiliation(s)
| | - Myung Ho Jeong
- Correspondence to: Myung Ho Jeong, M.D, Ph.D., FACC, FESC, FSCAI, Chief of Cardiovascular Medicine, Director of Cardiac Catheterization Laboratory, The Heart Center of Chonnam National University Hospital, Hakdong 8, Dong-gu, Gwangju, 501-757, Korea, Tel: 82-62-220-6243, Fax: 82-62-228-7174, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ott I, Vukovich R, Schömig A, Neumann FJ. Overexpression of glycosyl phosphatidylinositol-anchored tissue factor pathway inhibitor-1 inhibits tissue factor activity. Blood Coagul Fibrinolysis 2003; 14:539-44. [PMID: 12960606 DOI: 10.1097/00001721-200309000-00004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The cellular initiation of coagulation by the tissue factor (TF)-activated factor VII complex is transiently inhibited by endogenous tissue factor pathway inhibitor-1 (TFPI-1), whereas exogenously added TFPI-1 is targeted to a degradation pathway. This study investigates the relevance of glycosyl phosphatidylinositol (GPI) anchoring for the anticoagulant properties of TFPI-1. Experiments were performed with the human cell line ECV304 using liposomal gene transfer. For GPI anchoring of TFPI-1 we used a fusion protein of TFPI-1 and the GPI attachment sequence of decay-accelerating factor (GPI-TFPI-1), and compared it with wild-type TFPI-1. We measured TF and TFPI-1 surface expression by flow cytometry and TF proteolytic activity by a chromogenic assay for activated factor X generation. After transfection of GPI-TFPI-1, surface expression of TFPI-1 increased to 134 +/- 9% of mock transfected cells (mean +/- SEM, P = 0.004), and transfection with wild-type TFPI-1 did not significantly alter TFPI-1 surface expression. After transfection with GPI-TFPI-1, TF activity was reduced by 18 +/- 9% compared with mock transfections (P = 0.003), whereas after transfection with TFPI-1 wild type no significant inhibition was observed. This effect was not due to altered TF expression. GPI anchoring is an essential prerequisite for surface expression of TFPI-1 and inhibition of TF activity. Gene transfer of GPI-anchored TFPI, therefore, may be an efficient tool to inhibit local TF-induced coagulation.
Collapse
Affiliation(s)
- Ilka Ott
- Medizinische Klinik und Deutsches Herzzentrum, Technische Universität München, Lazarettstr. 36, 80636 München, Germany.
| | | | | | | |
Collapse
|
26
|
Chen X, Ren S, Ma MG, Dharmalingam S, Lu L, Xue M, Ducas J, Shen GX. Hirulog-like peptide reduces restenosis and expression of tissue factor and transforming growth factor-beta in carotid artery of atherosclerotic rabbits. Atherosclerosis 2003; 169:31-40. [PMID: 12860248 DOI: 10.1016/s0021-9150(03)00105-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Restenosis is responsible to approximately 30% of long-term failure following therapeutic vascular procedures. Thrombosis plays a key role in the development of restenosis. Thrombin-specific inhibitors have been considered as one type of candidates for the prevention of restenosis. Previous studies by our group demonstrated that a novel thrombin-specific inhibitor, hirulog-like peptide (HLP), reduced balloon catheter-induced neointima formation in rat carotid arteries. The present study examined the effect of HLP on angioplasty-induced restenosis in carotid arteries of atherosclerotic rabbits. New Zealand white rabbits were subject to air desiccation of the lumen of the right carotid arteries, then received high cholesterol/fat diet for 3 weeks. The rabbits were intravenously infused with HLP (1.6 mg/(kg/h)) or saline (n=7 per group) for 4 h started before angioplasty which dilated atherosclerotic lesions in right common carotid artery. Four weeks after the angioplasty, neointimal area, stenosis and neointima/media ratio in injured carotid arteries were reduced in atherosclerotic rabbits treated with HLP compared to saline controls by 62, 39 and 59% (P<0.05). The expression of tissue factor (TF) and transforming growth factor (TGF)-beta in the neointima of carotid arteries of rabbits treated with HLP was significantly weaker than saline controls (P<0.05 or <0.01). Activated partial thromboplastin time and bleeding time in HLP-treated rabbits were not significantly prolonged compared to controls. The results of the present study suggest that HLP may substantially reduce angioplasty-induced restenosis in atherosclerotic rabbits without increasing bleeding tendency. The inhibition on the expression of TF and TGF-beta in the neointima of the arterial wall may contribute to the preventive effect of HLP on restenosis in atherosclerotic rabbits.
Collapse
Affiliation(s)
- Xing Chen
- Department of Internal Medicine, University of Manitoba, Winnipeg, Man, Canada R3E 3P4
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abendschein DR, Yang LY, Chun J, Cho D, Scherrer D, St Pierre J. Prolonged procoagulant activity on overstretch-injured coronary arteries in pigs. J Thromb Haemost 2003; 1:836-42. [PMID: 12871423 DOI: 10.1046/j.1538-7836.2003.00100.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This study was designed to assess the time course and nature of the vascular procoagulant response after 1.5-fold balloon overstretch injury of the coronary arteries in pigs. Arteries were excised for chromogenic assay of bound factor (F)Xa and thrombin at 24 h, 3 days, 1 week, or 2 weeks after injury. FXa at the site of injury remained elevated for 1 week (4.9 +/- 5.9 microg cm(-2), n = 10), compared with non-injured control arteries (0.4 +/- 0.2 microg cm(-2), n = 18, P = 0.00025), while thrombin was increased only at 24 h. Tissue factor protein was abundant in non-injured coronaries (10 +/- 6 ng microg(-1) total protein, n = 9) and levels were unchanged by injury (13 +/- 11 ng microg(-1), n = 6) or 24-h administration of tissue factor pathway inhibitor (16 +/- 6 ng microg(-1), n = 6). Persistent tissue factor-mediated procoagulant activity may explain the need for prolonged anticoagulation to attenuate neointimal formation after balloon-induced coronary injury.
Collapse
Affiliation(s)
- D R Abendschein
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Shirotani-Ikejima H, Kokame K, Hamuro T, Bu G, Kato H, Miyata T. Tissue factor pathway inhibitor induces expression of JUNB and GADD45B mRNAs. Biochem Biophys Res Commun 2002; 299:847-52. [PMID: 12470656 DOI: 10.1016/s0006-291x(02)02759-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Tissue factor pathway inhibitor (TFPI) is a Kunitz-type protease inhibitor that regulates tissue factor-triggered blood coagulation. It has previously been reported that TFPI inhibits the proliferation of human umbilical vein endothelial cells (HUVECs), suggesting that TFPI may act as more than just a mediator of coagulation through changes in gene expression. By using DNA-array techniques and Northern blot analysis, we here revealed that TFPI transiently induced the mRNA expression of JUNB and GADD45B. The inducible effects were not observed in TFPIdeltaC (lacking the C-terminal basic region) or antithrombin (heparin-binding anticoagulant protease inhibitor). Moreover, the TFPI-induced expression of GADD45B was blocked by receptor-associated protein, which masks the ligand-binding domain of very low density lipoprotein receptor (VLDL-R). In conclusion, this is the first report to show an effect of TFPI on mRNA expression, and suggests that TFPI modulates cellular functions by inducing JUNB and GADD45B expression through binding to VLDL-R.
Collapse
Affiliation(s)
- Hiroko Shirotani-Ikejima
- National Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Golino P, Ragni M, Cimmino G, Forte L. Role of tissue factor pathway inhibitor in the regulation of tissue factor-dependent blood coagulation. CARDIOVASCULAR DRUG REVIEWS 2002; 20:67-80. [PMID: 12070535 DOI: 10.1111/j.1527-3466.2002.tb00083.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Tissue factor pathway inhibitor (TFPI) is a multivalent, Kunitz-type plasma proteinase inhibitor that modulates tissue factor-dependent coagulation in vivo. TFPI possesses a peculiar two-step mechanism of action; it directly inhibits activated factor X and subsequently produces feedback inhibition of the factor VIIa/tissue factor catalytic complex in a factor Xa-dependent fashion. TFPI biochemistry and physiology have been extensively studied during the last decade. Its pathophysiologic role in thrombotic disorders has, however, only recently started to be unraveled. In particular, circulating plasma TFPI levels have been found to modulate the activity of the tissue factor-dependent coagulation cascade. In animal models, neutralization of circulating TFPI activity results in restoration of intravascular thrombus formation previously abolished by aspirin. In patients with acute myocardial infarction, TFPI plasma levels measured in blood samples obtained from the coronary sinus were significantly lower than those measured in blood obtained from the ascending aorta, indicating acute consumption of TFPI within the coronary circulation of patients with intracoronary thrombosis. Finally, recent data indicate that transfection of the arterial wall with the gene coding for TFPI is an effective therapeutic intervention to prevent intravascular thrombus formation. Taken together, these observations underline the pathophysiologic importance of TFPI in regulating the procoagulant activity of tissue factor and open new potential therapeutic approaches for the treatment of thrombotic disorders.
Collapse
Affiliation(s)
- Paolo Golino
- Division of Cardiology, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy.
| | | | | | | |
Collapse
|
30
|
Abstract
It is widely accepted that blood coagulation in vivo is initiated during normal hemostasis, as well as during intravascular thrombus formation, when the cell-surface protein, tissue factor (TF), is exposed to the blood as a consequence of vascular injury. In addition to its essential role in hemostasis, tissue factor may be also implicated in several pathophysiological processes, such as intracellular signaling, cell proliferation, and inflammation. For these reasons, the tissue factor:factor VIIa complex has been the subject of intense research focus. Many experimental studies have demonstrated that inhibition of tissue factor:factor VIIa procoagulant activity are powerful inhibitors of in vivo thrombosis and that this approach usually results in less pronounced bleeding tendency, as compared to other "more classical" antithrombotic interventions. Alternative approaches may be represented by transfecting the arterial wall with natural inhibitors of tissue factor:factor VIIa complex, such as tissue factor pathway inhibitor (TFPI), which may result in complete inhibition of local thrombosis without incurring in potentially harmful systemic effects. Additional studies are warranted to determine the efficacy and safety of such approaches in patients.
Collapse
Affiliation(s)
- Paolo Golino
- Division of Cardiology, Seconda Università di Napoli, Piazza L Miraglia, 80138, Naples, Italy.
| |
Collapse
|
31
|
Abstract
Exposure of blood to tissue factor (TF) sets off the coagulation cascade. TF is a transmembrane protein that serves as an essential cofactor for activated coagulation factor VII (FVIIa). TF may be exposed locally by vascular injury (such as balloon angioplasty) or by spontaneous rupture of an atherosclerotic plaque. Expression of TF may also be induced on monocytes and endothelial cells in conditions like sepsis and cancer, causing a more generalised activation of clotting. TF may thus play a central role in thrombosis in a number of settings, and attention has turned to blocking TF as a means to prevent thrombosis. Inhibiting the inducible expression of TF by monocytes can be achieved by 'deactivating' cytokines, such as interleukin (IL)-4, -10 and -13, or by certain prostanoids; by drugs that modify signal transduction, such as pentoxifylline, retinoic acid or vitamin D(3), or by antisense oligonucleotides. Such approaches are for the most part at a preclinical stage. The function of TF can be blocked by antibodies that prevent the binding of FVIIa to TF; by active site-inhibited FVIIa, which competes with native FVIIa for binding; by antibodies or small molecules that block the function of the TF/FVIIa complex; and by molecules, such as TF pathway inhibitor or nematode anticoagulant peptide C2, which inhibit the active site of FVIIa in the TF/FVIIa complex after first binding to activated factor X. The latter two agents have entered Phase II clinical trials. Perhaps most intriguing is the use of anti-TF agents locally, which holds the promise of stopping thrombosis at a specific site of injury without the bleeding risk associated with systemic anticoagulation.
Collapse
Affiliation(s)
- Donald S Houston
- Section of Hematology/Oncology, Department of Internal Medicine, University of Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, R3E 0V9, Canada.
| |
Collapse
|
32
|
Kaiser B, Hoppensteadt DA, Fareed J. Tissue factor pathway inhibitor: an update of potential implications in the treatment of cardiovascular disorders. Expert Opin Investig Drugs 2001; 10:1925-35. [PMID: 11772296 DOI: 10.1517/13543784.10.11.1925] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tissue factor (TF) plays a crucial role in the pathogenesis of thrombotic, vascular and inflammatory disorders. Thus, the inhibition of this membrane protein provides a unique therapeutic approach for prophylaxis and/or treatment of various diseases. Tissue factor pathway inhibitor (TFPI), the only endogenous inhibitor of the TF/Factor VIIa (FVIIa) complex, has recently been characterised biochemically and pharmacologically. Studies in patients demonstrated that both TF and TFPI may be indicators for the course and the outcome of cardiovascular and other diseases. Based on experimental and clinical data, TFPI might become an important drug for several clinical indications. TFPI is expected to inhibit the development of post-injury intimal hyperplasia and thrombotic occlusion in atherosclerotic vessels as well as to be effective in acute coronary syndromes, such as unstable angina and myocardial infarction. Of special interest is the inhibition of TF-mediated processes in sepsis and disseminated intravascular coagulation (DIC), which are associated with the activation of various inflammatory pathways as well as of the coagulation system. A Phase II trial of the efficacy of TFPI in patients with severe sepsis showed a mortality reduction in TFPI- compared to placebo-treated patients and an improvement of organ dysfunctions. TFPI can be administered exogenously in high doses to suppress TF-mediated effects, alternatively high amounts of TFPI can be released from intravascular stores by other drugs, such as heparin and low molecular weight heparins (LMWH). Using this method high concentrations of the inhibitor are provided at sites of tissue damage and ongoing thrombosis. At present, clinical studies with TFPI are rather limited so that the clinical potential of the drug cannot be assessed properly. However, TFPI and its variants are expected to undergo further development and to find indications in various clinical states.
Collapse
Affiliation(s)
- B Kaiser
- Friedrich Schiller University Jena,Center for Vascular Biology and Medicine,Nordhäuser Str. 78, D-99089 Erfurt, Germany.
| | | | | |
Collapse
|
33
|
Hathcock JJ, Hall CL, Turitto VT. Active tissue factor shed from human arterial smooth muscle cells adheres to artificial surfaces. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2001; 11:1211-25. [PMID: 11263809 DOI: 10.1163/156856200744282] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Through a series of in vitro assays, this study outlines a flow-mediated process by which active tissue factor (TF), the prime initiator of coagulation, may be transferred from the plasma membrane of vascular smooth muscle cells (VSMCs) to that of artificial surfaces such as those typically associated with intravascular implants. Studies with quiescent and activated rat VSMCs demonstrated that pathologically high shear stresses (tau(w) = 250 dyn cm(-2)) resulted in the loss of TF activity from the cell surface. Subsequent experiments with human VSMCs showed that VSMCs continuously release active TF into their extracellular medium, presumably in the form of lipid vesicles or microparticles, and that fluid shear stress (tauw = 50 dyncm(-2)) or chemical agonists (A23187) can significantly accelerate this release. Experiments with a wide array of polymeric and metallic materials showed that the TF shed from VSMCs was able to adhere to these surfaces and promote the activation of coagulation factor X (FX) at the material surface. Extracellular TF bound strongly to both uncoated and human plasma coated surfaces under a wide range of hemodynamic shear stresses (0-20 dyncm(-2)). When an extracellular, VSMC-derived TF mixture was perfused over Ti 6-4 surfaces, the adhesion of TF was found to be time-dependent, gradually accumulating on the material surface over time. Thus an important criterion in the design or success of intravascular devices may be related to their ability to interact with TF, shed from cell surfaces. This is especially important as TF may lead to thrombotic complications, the products of which may also increase cellular proliferation.
Collapse
Affiliation(s)
- J J Hathcock
- Department of Biomedical Engineering, The University of Memphis, TN 38152, USA.
| | | | | |
Collapse
|
34
|
Golino P, Cirillo P, Calabro' P, Ragni M, D'Andrea D, Avvedimento EV, Vigorito F, Corcione N, Loffredo F, Chiariello M. Expression of exogenous tissue factor pathway inhibitor in vivo suppresses thrombus formation in injured rabbit carotid arteries. J Am Coll Cardiol 2001; 38:569-76. [PMID: 11499754 DOI: 10.1016/s0735-1097(01)01350-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVES The aim of the present study was to test the hypothesis that retrovirus-mediated in vivo tissue factor pathway inhibitor (TFPI) gene transfer to the arterial wall would efficiently inhibit thrombosis without causing significant changes in systemic hemostatic variables. BACKGROUND Acute coronary syndromes (unstable angina and acute myocardial infarction) are usually caused by atherosclerotic plaque rupture, with consequent activation of the coagulation cascade and circulating platelets. Tissue factor (TF) exposure represents an early event in this pathophysiologic sequence, leading to activation of the extrinsic coagulation pathway and thrombin formation. Tissue factor pathway inhibitor is a naturally occurring inhibitor of the extrinsic pathway. METHODS In the present study, the gene coding for rabbit TFPI was inserted in a retroviral vector under control of a tetracycline-inducible promoter. Replication-defective, infectious, recombinant retroviruses were used to transfect rabbit carotid arteries with either TFPI or a reporter gene--green fluorescent protein (GFP). RESULTS Retroviral-mediated arterial gene transfer of TFPI resulted in potent inhibition of intravascular thrombus formation in stenotic and injured rabbit carotid arteries, whereas transfection of the contralateral carotid artery with GFP had no effect on thrombosis. No significant changes in systemic hemostatic variables (prothrombin time and partial thromboplastin time) were observed when thrombosis was inhibited. CONCLUSIONS These data suggest that retroviral-mediated transfection of the arterial wall with TFPI might represent an attractive approach for the treatment of thrombotic disorders.
Collapse
Affiliation(s)
- P Golino
- Department of Internal Medicine, University of Naples Federico II, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Targeted contrast agents are expanding the detectability and diagnosis of pathology from a strict anatomic to biochemical basis. Moreover, these new agents, in their various forms, offer the potential for site-specific drug and gene delivery, ie, the "magic bullet" first postulated by Paul Erhlich 100 years ago. The ability to direct drugs to the molecular signatures of disease, to confirm noninvasively their presence at the site-of-interest, and to quantify the adequacy of local drug concentration at the time of treatment, ie, rational targeted drug delivery, offers exciting new clinical paradigms in the near future.
Collapse
Affiliation(s)
- G M Lanza
- Division of Cardiology, Washington University Medical School, St Louis, MO 63110, USA
| | | |
Collapse
|
36
|
Sun LB, Utoh J, Moriyama S, Tagami H, Okamoto K, Kitamura N. Topically applied tissue factor pathway inhibitor reduced intimal thickness of small arterial autografts in rabbits. J Vasc Surg 2001; 34:151-5. [PMID: 11436089 DOI: 10.1067/mva.2001.113307] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
PURPOSE The purpose of this study was to investigate whether topically applied tissue factor pathway inhibitor (TFPI) reduces intimal thickness and increases long-term patency of small arterial autografts in rabbits. METHODS An entire 10-mm long section of the left femoral artery was harvested and immersed in saline solution (control group, n = 10), 100 IU/mL of heparin (heparin group, n = 15), or 40 microg/mL of TFPI (TFPI group, n = 15) for 15 minutes. Then the graft was interposed to the right femoral artery. Patency rates were determined by flow measurements throughout the time course of the study, and the grafts were analyzed for measurement of intimal thickness at 3 months after operation. Immunohistochemical analysis was performed to examine whether topically applied TFPI binds to endothelial cells of the grafts. RESULTS Three-month postoperative patency rates were 10% in the control group, 47% in the heparin group, and 73% in the TFPI group. The TFPI group had a significantly higher patency rate than that of the control group (P <.005). Compared with the heparin group, the TFPI group had a significant reduction in intimal area (0.19 +/- 0.05 mm(2) vs 0.30 +/- 0.09 mm(2), P =.0051), in percentage of stenosis (35.7% +/- 7.7% vs 61.4% +/- 15.8%, P <.0001), and in intimal/media areas ratio (0.64 +/- 0.24 vs. 1.04 +/- 0.33, P =.0051). Immunohistologic analyses confirmed that topically applied TFPI bound to endothelial cells. CONCLUSION These results indicate that topically applied TFPI reduces intimal thickness and increases long-term patency of small arterial autografts in rabbits.
Collapse
Affiliation(s)
- L B Sun
- First Department of Surgery, Kumamoto University School of Medicine, Japan.
| | | | | | | | | | | |
Collapse
|
37
|
Neumann F, Kastrati A, Miethke T, Pogatsa-Murray G, Mehilli J, Valina C, Jogethaei N, da Costa CP, Wagner H, Schömig A. Treatment of Chlamydia pneumoniae infection with roxithromycin and effect on neointima proliferation after coronary stent placement (ISAR-3): a randomised, double-blind, placebo-controlled trial. Lancet 2001; 357:2085-9. [PMID: 11445102 DOI: 10.1016/s0140-6736(00)05181-3] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Vascular infection with Chlamydia pneumoniae might boost inflammatory responses that play a pivotal part in neointima formation, which is the main cause of restenosis after stenting. Our aim was to investigate whether or not treatment of C pneumoniae infection with antibiotics prevents restenosis after coronary stent placement. METHODS We enrolled 1010 consecutive patients with successful coronary stenting into a randomised, double-blind trial. Patients received the macrolide antibiotic roxithromycin 300 mg once daily for 28 days (506), or placebo (504). Primary endpoint was frequency of restenosis (diameter stenosis >50%) at follow-up angiography, and secondary endpoint was rate of target vessel revascularisation during the year after stenting. A prespecified secondary analysis addressed treatment effect with respect to titre of C pneumoniae in serum. Analysis was by intention to treat. FINDINGS Rate of angiographic restenosis was 31% (157 lesions) in the roxithromycin group and 29% (148) in the placebo group (relative risk 1.08 [95% CI 0.92-1.26]; p50.43), corresponding to a rate of target vessel revascularisation of 19% (120) and 17% (105), respectively (1.13 [0.95-1.36]; p50.30). The combined 1-year rates of death and myocardial infarction were 7% (36) in the roxithromycin group and 6% (30) in the placebo group (p50.45). We showed a significant interaction between treatment and C pneumoniae antibody titre (p50.038 for restenosis, p50.006 for revascularisation), favouring roxithromycin at high titres (adjusted odds ratios at a titre of 1/512 were 0.44 [0.19-1.06] and 0.32 [0.13-0.81], respectively). INTERPRETATION Non-selective use of roxithromycin is inadequate for prevention of restenosis after coronary stenting. There is, however, a differential effect dependent on C pneumoniae titres. In patients with high titres, roxithromycin reduced the rate of restenosis.
Collapse
Affiliation(s)
- F Neumann
- Medizinische Klinik, Technische Universität München, 81675, München, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Moatti D, Meirhaeghe A, Ollivier V, Bauters C, Amouyel P, de Prost D. Polymorphisms of the tissue factor pathway inhibitor gene and the risk of restenosis after coronary angioplasty. Blood Coagul Fibrinolysis 2001; 12:317-23. [PMID: 11460016 DOI: 10.1097/00001721-200106000-00013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Intracoronary stent implantation is associated with a significantly lower risk of restenosis compared with balloon angioplasty. However, restenosis still occurs in some cases. Experimental studies suggest that the tissue factor pathway is involved in this phenomenon. We investigated a possible relationship between three previously identified polymorphisms of the tissue factor pathway inhibitor (TFPI) gene and restenosis in 443 patients who underwent angioplasty, with or without stent implantation. The effect of the intron 7-33T<--C polymorphism and that of the combined intron 7 and promoter genotype on plasma TFPI levels was also investigated in 58 healthy subjects. DNA analysis was performed by polymerase chain reaction amplification of genomic DNA extracted from white blood cells, followed by digestion with the restriction enzymes Hind III, Nde I and Mae III for the detection of promoter, intron 7 and exon IX polymorphisms, respectively. The minimal luminal diameter, percent stenosis, acute gain, late loss and loss index did not differ according to the genotype before, immediately after or 6 months after angioplasty, regardless of stent implantation. Interestingly, subjects with the intron 7 CC genotype had significantly higher total TFPI levels than those with the TT genotype before and after an enoxaparin injection. Moreover, subjects with the -287TT/Int7TT combined genotype had the lowest plasma TFPI levels. Despite significant variations in plasma TFPI levels, we found no evidence that three polymorphisms of the TFPI gene influence the risk of restenosis. These results do not exclude the possibility that other polymorphisms in the TFPI gene may influence this risk.
Collapse
Affiliation(s)
- D Moatti
- INSERM U479, Faculté Xavier Bichat, Paris, France
| | | | | | | | | | | |
Collapse
|
39
|
Söderström T, Hedner U, Arnljots B. Active site-inactivated factor VIIa prevents thrombosis without increased surgical bleeding: topical and intravenous administration in a rat model of deep arterial injury. J Vasc Surg 2001; 33:1072-9. [PMID: 11331851 DOI: 10.1067/mva.2001.112808] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
PURPOSE The primary event in the procoagulant response after vascular interventions is the tissue factor (TF)-factor VIIa complex formation, which occurs when TF is exposed to the circulating blood by the inflicted trauma. Human recombinant active site-inhibited coagulation factor VIIa (FFR-rFVIIa) binds well to TF but cannot initiate blood coagulation, and should thereby block thrombus formation. This hypothesis was tested with a rat model of arterial thrombosis. METHODS In a blinded randomized study, the antithrombotic and antihemostatic effects of FFR-rFVIIa and heparin were evaluated in a rat model of mechanical deep arterial injury. In one arm of the study, FFR-rFVIIa (0.2 mg in 150 microL) or vehicle alone was applied topically at the site of vascular injury. In the other arm, FFR-rFVIIa (4 mg/kg), heparin (1 mg/kg), or vehicle alone was injected intravenously. RESULTS FFR-rFVIIa produced a powerful antithrombotic effect after both topical and intravenous administrations (P =.02 and P =.005, respectively) without increasing the surgical bleeding. Heparin prevented thrombosis equally well as FFR-rFVIIa (P =.0007), but doubled the surgical bleeding compared with FFR-rFVIIa (P =.03) and controls (P =.008). In the topical study, the antithrombotic effect was achieved without altering parameters of plasma anticoagulation (prothrombin time and activated partial thromboplastin time) or producing detectable levels of FFR-rFVIIa in plasma. CONCLUSION In this model FFR-rFVIIa effectively inhibits thrombus formation without the expense of increased surgical bleeding, which indicates the potential of FFR-rFVIIa as an effective and safe strategy for prevention of thrombosis in reconstructive vascular surgery and various forms of percutaneous revascularization.
Collapse
Affiliation(s)
- T Söderström
- Department of Plastic and Reconstructive Surgery, University Hospital, Malmö, Sweden
| | | | | |
Collapse
|
40
|
Zoldhelyi P, Chen ZQ, Shelat HS, McNatt JM, Willerson JT. Local gene transfer of tissue factor pathway inhibitor regulates intimal hyperplasia in atherosclerotic arteries. Proc Natl Acad Sci U S A 2001; 98:4078-83. [PMID: 11274432 PMCID: PMC31182 DOI: 10.1073/pnas.061004098] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tissue factor (TF), the initiator of blood coagulation and thrombosis, is up-regulated after vascular injury and in atherosclerotic states. Systemic administration of recombinant TF pathway inhibitor (TFPI) has been reported to decrease intimal hyperplasia after vascular injury and also to suppress systemic mechanisms of blood coagulation and thrombosis. Here we report that, in heritable hyperlipidemic Watanabe rabbits, adenoviral gene transfer of TFPI to balloon-injured atherosclerotic arteries reduced the extent of intimal hyperplasia by 43% (P < 0.05) compared with a control vector used at identical titer (1 x 10(10) plaque-forming units/ml). Platelet aggregation and coagulation studies performed 7 days after local gene transfer of TFPI failed to show any impairment in systemic hemostasis. At time of sacrifice, 4 weeks after vascular injury, the 10 Ad-TFPI treated carotid arteries were free of thrombi, whereas two control-treated arteries were occluded (P, not significant). These findings suggest that TFPI overexpressed in atherosclerotic arteries can regulate hyperplastic response to injury in the absence of changes in the hemostatic system, establishing a role for local TF regulation as target for gene transfer-based antirestenosis therapies.
Collapse
Affiliation(s)
- P Zoldhelyi
- Wafic Said Molecular Cardiology and Gene Therapy Research Laboratory, Texas Heart Institute, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
41
|
|
42
|
Huynh TT, Davies MG, Thompson MA, Ezekowitz MD, Hagen P, Annex BH. Local treatment with recombinant tissue factor pathway inhibitor reduces the development of intimal hyperplasia in experimental vein grafts. J Vasc Surg 2001; 33:400-7. [PMID: 11174796 DOI: 10.1067/mva.2001.111989] [Citation(s) in RCA: 25] [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
BACKGROUND Tissue factor (TF)-initiated thrombin generation has been implicated in the development of intimal hyperplasia after arterial injury. An increase in intimal TF expression has been shown to precede the development of intimal hyperplasia in vein grafts. This study examines the effects of local treatment with recombinant human tissue factor pathway inhibitor (rTFPI) in experimental vein grafts. METHODS Thirty-six male New Zealand white rabbits underwent bypass grafting of the carotid artery by use of the reversed ipsilateral jugular vein and were divided into four groups. Twenty animals had ex vivo incubation with rTFPI treatment (50 microg x mL(-1); n = 10) or placebo vehicle (control; n = 10). Sixteen animals received both ex vivo incubation and in vivo gel treatment with rTFPI (50 microg. mL(-1); n = 8) or without rTFPI (gel-control; n = 8). After operation, vein grafts were harvested at 3 days for immunohistochemical and Western analyses and at 28 days for histomorphologic study. RESULTS Western analysis demonstrated a 6.2-fold reduction in the expression of TF protein with rTFPI treatment in comparison to without rTFPI treatment. CD-18 leukocyte staining was diminished, whereas Tie-2 endothelial staining was increased in all rTFPI-treated vein grafts, compared with control and gel-control vein grafts. Intimal thickness was reduced by 21% with ex vivo rTFPI treatment compared with placebo (69 +/- 4 versus 87 +/- 5 microm; P <.05) and by 30% with the addition of rTFPI in vivo compared with gel-control (60 +/- 4 versus 86 +/- 5 microm; P <.01). CONCLUSION Local administration of rTFPI exerts early beneficial effects and limits the development of intimal hyperplasia in vein grafts. Therefore blocking TF-mediated pathway may offer new therapeutic options to reduce vein graft failure.
Collapse
Affiliation(s)
- T T Huynh
- Department of Surgery, Duke University Medical Center and Durham VA Medical Center, NC, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Roqué M, Reis ED, Fuster V, Padurean A, Fallon JT, Taubman MB, Chesebro JH, Badimon JJ. Inhibition of tissue factor reduces thrombus formation and intimal hyperplasia after porcine coronary angioplasty. J Am Coll Cardiol 2000; 36:2303-10. [PMID: 11127477 DOI: 10.1016/s0735-1097(00)01018-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVES We investigated the in vivo effects of tissue factor (TF) inhibition with recombinant tissue factor pathway inhibitor (rTFPI) on acute thrombus formation and intimal hyperplasia and the in vitro effects on smooth muscle cell migration and proliferation. BACKGROUND Inhibition of TF with TFPI has been shown to reduce intimal hyperplasia in experimental models. However, its effects after coronary angioplasty and the cellular mechanisms involved have not been investigated. METHODS Twenty-three swine underwent multivessel coronary angioplasty. Fifteen (n = 25 arteries) were euthanized at 72 h to assess thrombus formation and eight (n = 24 arteries) at 28 days to assess intimal hyperplasia. Animals in the 72-h time point received: 1) human rTFPI (0.5 mg bolus plus 25 microg/kg/min continuous infusion for 3 days) plus heparin (150 IU/kg intravenous bolus) plus acetyl salicylic acid (ASA) (325 mg/day); 2) rTFPI regimen plus ASA and 3) heparin (150 IU/kg intravenous bolus) plus ASA. RESULTS On histology the control group had evidence of mural thrombus (area 0.8+/-0.4 mm2). Treatment with TFPI plus heparin abolished thrombus formation (mean area: 0.0+/-0.0 mm2, p < 0.05) but was associated with prolonged activated partial thromboplastin time and extravascular hemorrhage. Recombinant TFPI alone inhibited thrombosis without bleeding complications (mean area: 0.03+/-0.02 mm2, p < 0.05 vs. control). Animals in the 28-day time point received continuous intravenous infusion of rTFPI or control solution for 14 days. Tissue factor pathway inhibitor reduced neointimal formation with mean intimal area of 1.2+/-0.3 mm2 versus 3.2+/-0.4 mm2 in the control group; p < 0.01. Recombinant TFPI had no effect on human aortic smooth muscle cell growth but inhibited platelet-derived growth factor BB-induced migration. CONCLUSIONS Inhibition of TF with rTFPI can prevent acute thrombosis and intimal hyperplasia after injury. Tissue factor plasma inhibitor may prove useful as an adjunct to intracoronary interventions.
Collapse
Affiliation(s)
- M Roqué
- Zena and Michael A. Wiener Cardiovascular Institute, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lanza GM, Abendschein DR, Hall CS, Scott MJ, Scherrer DE, Houseman A, Miller JG, Wickline SA. In vivo molecular imaging of stretch-induced tissue factor in carotid arteries with ligand-targeted nanoparticles. J Am Soc Echocardiogr 2000; 13:608-14. [PMID: 10849515 DOI: 10.1067/mje.2000.105840] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Molecular imaging permits tissues to be functionally characterized by identification of specific cell-surface receptors with targeted contrast agents. In our study, a ligand-targeted acoustic nanoparticle system was used to identify the angioplasty-induced expression of tissue factor by smooth muscle cells within the tunica media. Pig carotid arteries were overstretched bilaterally with balloon catheters, treated with a tissue factor-targeted or a control nanoparticle system, and imaged with intravascular ultrasound (20 MHz) before and after treatment. Carotid wall acoustic reflectivities were unaffected by overstretch injury. Tissue factor-targeted nanoemulsion bound and increased the echogenicity of smooth muscle cells expressing tissue factor within the tunica media. The targeted emulsion increased the arterial wall gray scale (99.4+/-14.5; P<.05) relative to pretreatment (41.8+/-11.1, P<0.05) and the control gray scale (pre-emulsion: 49.3+/-9.5; post-emulsion: 43.7+/-6.4; P<.05). The area of acoustic enhancement appeared to coincide with expression of induced tissue factor in the tunica media confirmed by immunohistochemistry. We have demonstrated that this novel nanoemulsion can infiltrate into arterial walls after balloon injury and localize the expression of overstretch-induced tissue factor within pig carotid arteries. Molecular imaging and quantification of complex, biochemical change, such as tissue factor expression after angioplasty, may prove to be a prognostically important predictor of subsequent restenosis.
Collapse
Affiliation(s)
- G M Lanza
- Department of Medicine, Division of Cardiology, Barnes-Jewish Hospital, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Lanza GM, Abendschein DR, Hall CS, Marsh JN, Scott MJ, Scherrer DE, Wickline SA. Molecular imaging of stretch-induced tissue factor expression in carotid arteries with intravascular ultrasound. Invest Radiol 2000; 35:227-34. [PMID: 10764091 DOI: 10.1097/00004424-200004000-00003] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
RATIONALE AND OBJECTIVES Molecular imaging with targeted contrast agents enables tissues to be distinguished by detecting specific cell-surface receptors. In the present study, a ligand-targeted acoustic nanoparticle system is used to identify angioplasty-induced expression of tissue factor by smooth muscle cells within carotid arteries. METHODS Pig carotid arteries were overstretched with balloon catheters, treated with tissue factor-targeted or a control nanoparticle system, and imaged with intravascular ultrasound before and after treatment. RESULTS Tissue factor-targeted emulsions bound and increased the echogenicity and gray-scale levels of overstretched smooth muscle cells within the tunica media, versus no change in contralateral control arteries. Expression of stretch-induced tissue factor in carotid artery media was confirmed by immunohistochemistry. CONCLUSIONS The potential for abnormal thrombogenicity of balloon-injured arteries, as reflected by smooth muscle expression of tissue factor, was imaged using a novel, targeted, nanoparticulate ultrasonic contrast agent.
Collapse
Affiliation(s)
- G M Lanza
- Department of Medicine, Washington University Medical School, Barnes-Jewish Hospital, St. Louis, Missouri 63110, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Mizuno O, Hojo Y, Ikeda U, Katsuki T, Fukazawa H, Kurosaki K, Fujikawa H, Shimada K. Assessment of coagulation and platelet activation in coronary sinus blood induced by transcatheter coronary intervention for narrowing of the left anterior descending coronary artery. Am J Cardiol 2000; 85:154-60. [PMID: 10955369 DOI: 10.1016/s0002-9149(99)00651-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Influences of recently developed methods for coronary intervention on hemostasis in the coronary circulation are unclear. The objective of this study was to investigate changes in coagulation and platelet activation in the coronary circulation induced by percutaneous transluminal coronary angioplasty (PTCA). We studied 35 patients with coronary heart disease who underwent elective PTCA to isolated stenotic narrowing of left coronary arteries. Seven patients received only PTCA, 12 underwent percutaneous transluminal rotational atherectomy (PTRA), and 16 underwent stent implantation. Blood samples were drawn from the coronary sinus immediately before and after as well as 4 and 24 hours after PTCA. Plasma levels of tissue factor (TF), thrombin-antithrombin III complex, plasminogen activator inhibitor (PAI)-1, tissue plasminogen activator (t-PA), beta-thromboglobulin, and platelet factor 4 were measured by enzyme-linked immunosorbent assay. In all patients, TF levels in the coronary sinus blood showed significant increases 4 and 24 hours after PTCA and thrombin-antithrombin III complex levels showed significant increases 24 hours after PTCA. PAI-1 showed significant increases 24 hours after PTCA and t-PA showed significant increases 4 and 24 hours after PTCA. Changes in levels of these markers by PTCA were similar among the 3 groups. In PTRA, levels of beta-thromboglobulin and platelet factor 4, markers of platelet activation, increased immediately after the procedure and returned to baseline levels after 4 hours. PTCA induced increases in blood coagulation and fibrinolysis in the coronary circulation. PTRA caused a marked but transient activation of platelets. These changes may contribute to acute complications during the procedure.
Collapse
Affiliation(s)
- O Mizuno
- Department of Cardiology, Jichi Medical School, Tochigi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Soejima H, Ogawa H, Yasue H, Kaikita K, Takazoe K, Nishiyama K, Misumi K, Miyamoto S, Yoshimura M, Kugiyama K, Nakamura S, Tsuji I. Angiotensin-converting enzyme inhibition reduces monocyte chemoattractant protein-1 and tissue factor levels in patients with myocardial infarction. J Am Coll Cardiol 1999; 34:983-8. [PMID: 10520779 DOI: 10.1016/s0735-1097(99)00318-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES We investigated the effects of enalapril therapy on plasma tissue factor (TF), tissue factor pathway inhibitor (TFPI) and monocyte chemoattractant protein-1 (MCP-1) levels in patients with acute myocardial infarction. BACKGROUND Macrophages express TF in human coronary atherosclerotic plaques. Both TF and TFPI are major regulators of coagulation and thrombosis. Monocyte chemoattractant protein-1 is a monocyte and macrophage chemotactic and activating factor. METHODS In a randomized, double-blind, placebo-controlled study beginning about two weeks after myocardial infarction, 16 patients received four weeks of placebo (placebo group) and another 16 patients received four weeks of enalapril 5 mg daily therapy (enalapril group). We performed blood sampling after administration of the doses. RESULTS There were no significant differences in the serum angiotensin-converting enzyme (ACE) activity, plasma TF, free TFPI or MCP-1 levels before administration between the enalapril and placebo groups. In the enalapril group, ACE activity (IU/liter) (14.0 before, 5.2 on day 3, 5.8 on day 7, 6.3 on day 28), TF levels (pg/ml) (223, 203, 182, 178) and MCP-1 levels (pg/ml) (919, 789, 790, 803) significantly decreased by day 28. However, the free TFPI levels (ng/ml) (28.2, 26.5, 26.8, 28.4) did not change. These four variables were unchanged during the study period in the placebo group. CONCLUSIONS This study demonstrated that administration of enalapril reduces the increased procoagulant activity in patients with myocardial infarction associated with inhibition of the activation and accumulation of macrophages and monocytes.
Collapse
Affiliation(s)
- H Soejima
- Department of Cardiovascular Medicine, Kumamoto University School of Medicine, Kumamoto City, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Thrombosis is a key feature of the initiation and progression of atherosclerosis and its clinical sequelae. Acute thrombosis can lead to arterial occlusion and consequently provoke myocardial infarction, unstable angina, stroke and sudden death. Acute thrombosis can also be a complication of arterial bypass surgery, balloon angioplasty, atherectomy, or coronary artery stenting. The thrombotic response is influenced by several factors, among them the thrombogenicity of the vessel wall and of certain blood components as well as their interaction with the lipid pool. Tissue factor (TF) is considered to be the primary cofactor of cellular origin that is involved in activation of the coagulation pathway. The active form of TF has been shown to be present in specimens of human coronary artery in association both with acellular lipid areas and with macrophages and smooth muscle cells, which suggests that TF plays a major role in determining plaque thrombogenicity. We discuss here what is currently known about the role of tissue factor in atherogenesis, and focus attention on pharmacological approaches in this area.
Collapse
Affiliation(s)
- E Tremoli
- Institute of Pharmacological Sciences, E. Grossi Paoletti Center, University of Milan, Italy.
| | | | | | | |
Collapse
|
49
|
Visconti LO, Pezzano A, Quattrocchi G, Pezzano A. Procoagulant activity during coronary interventions and coronary artery patency. Int J Cardiol 1999; 68 Suppl 1:S23-7. [PMID: 10328607 DOI: 10.1016/s0167-5273(98)00300-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- L O Visconti
- II Divisione, Cardiologica De Gasperis, Ospedale Niguarda, Milano, Italy
| | | | | | | |
Collapse
|
50
|
Shinoda E, Yui Y, Hattori R, Tanaka M, Inoue R, Aoyama T, Takimoto Y, Mitsui Y, Miyahara K, Shizuta Y, Sasayama S. Tissue factor pathway inhibitor-2 is a novel mitogen for vascular smooth muscle cells. J Biol Chem 1999; 274:5379-84. [PMID: 10026147 DOI: 10.1074/jbc.274.9.5379] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A mitogen for growth-arrested cultured bovine aortic smooth muscle cells was purified to homogeneity from the supernatant of cultured human umbilical vein endothelial cells by heparin affinity chromatography and reverse-phase high performance liquid chromatography. This mitogen was revealed to be tissue factor pathway inhibitor-2 (TFPI-2), which is a Kunitz-type serine protease inhibitor. TFPI-2 was expressed in baby hamster kidney cells using a mammalian expression vector. Recombinant TFPI-2 (rTFPI-2) stimulated DNA synthesis and cell proliferation in a dose-dependent manner (1-500 nM). rTFPI-2 activated mitogen-activated protein kinase (MAPK) activity and stimulated early proto-oncogene c-fos mRNA expression in smooth muscle cells. MAPK, c-fos expression and the mitogenic activity were inhibited by a specific inhibitor of MAPK kinase, PD098059. Thus, the mitogenic function of rTFPI-2 is considered to be mediated through MAPK pathway. TFPI has been reported to exhibit antiproliferative action after vascular smooth muscle injury in addition to the ability to inhibit activation of the extrinsic coagulation cascade. However, structurally similar TFPI-2 was found to have a mitogenic activity for the smooth muscle cell.
Collapse
Affiliation(s)
- E Shinoda
- Third Division, Department of Internal Medicine, Kyoto University Hospital, Shogoin-Kawaracho 54, Kyoto 6068507, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|