Skip main navigation

The Intima

Soil for Atherosclerosis and Restenosis
Originally publishedhttps://doi.org/10.1161/01.RES.77.3.445Circulation Research. 1995;77:445–465

    Thirty years ago, John French wrote a seminal review describing the unique properties of the arterial intima.1 His major point was that the smooth muscle cells of the intima have a unique morphology (Fig 1). French also pointed out that intimal formation appeared during normal development and aging as well as in the response of arteries to almost any imaginable injury, including atherosclerosis.

    This article attempts to update French’s review. We will discuss the developmental origins of the intima and suggest that the arterial intima is a distinct tissue with a long and rapidly increasing list of differentially expressed genes (Table 1). This pattern of intimal gene expression may be responsible for the origins of atherosclerosis.

    The bulk of the review, however, deals with the posited role of smooth muscle replication in hypertension, atherosclerosis, and restenosis. Topics covered will include a revisiting of the 20-year-old observation that atherosclerosis is monoclonal2345 as well as reviews of current knowledge of the pharmacology of smooth muscle proliferation. Perhaps most important, however, we will present a critical discussion of evidence for and against a role for smooth muscle replication in these diseases.

    Formation of the Intima

    Surprisingly little is known about how vessels acquire a coat of smooth muscle cells. In the rat, the precursors of the carotid arteries lack a smooth muscle layer as late as day 12 of gestation.6 The first smooth muscle cells appear as a condensation around these endothelial tubes over a period of ≈24 hours. As the mesenchymal cells invest the endothelial tube, the cells begin to express smooth muscle α-actin.7 Smooth muscle α-actin, however, is not a definitive marker of smooth muscle lineage and can be found in many non–smooth muscle cell types.8 More definitive smooth muscle–specific proteins are expressed later in development. These include desmin, calponin, smooth muscle γ-actin, and the smooth muscle myosins.910111213 Whereas these latter genes seem largely restricted to smooth muscle cells, their expression is typically lost in cultured cells or in cells that form the neointima.

    It is important to realize that not all species go on to form an intima unless their arteries are injured. In humans, the intima develops spontaneously after birth14151617 and increases rapidly until ≈6 months of age.1417 French suggests that arterial intimal formation is a function of animal size, because arteries of smaller animals do not form an intima unless the vessel first undergoes trauma.1 The most interesting site of intimal growth is the origin of the left anterior descending coronary artery, a site with a high probability of developing atherosclerosis in later life.

    The only site where intimal formation has been well studied is the ductus arteriosus.1819 Contrary to conventional wisdom, this intima forms spontaneously before birth, not as the result of trauma associated with ductus closure. Therefore, it is intriguing to consider ductus closure as a model for diseases of the intima, including atherosclerosis and restenosis.

    Neointimal Formation: A Generic Response of Vessels to Injury?

    “Neointima” is an intima that forms in response to injury. We recognize a new intima because many arteries, especially smaller ones, either do not form an intima at all or do so only slowly as the animal ages.120 Neointimal formation, however, occurs in all arteries as a response to a wide variety of injuries, including radiation, application of turpentine to the adventitia, wrapping the vessel, and electrical stimulation, as well as mechanical injuries, including placement of a suture, scratching with a probe, or dilatation of the common carotid artery with an embolectomy balloon catheter.121222324252627282930 These changes are even seen in transplanted veins that undergo arterialization. Put another way, neointimal formation is as characteristic of responses to injury in the smooth muscle–rich vessel wall as fibrosis is characteristic of responses in the fibroblastic dermis of skin or gliosis is characteristic of the brain with its glial cells. The most obvious questions about neointimal formation are as follows: (1) Are intimal or neointimal cells, like glia, a distinct cell type? (2) What molecules control neointimal formation? (3) What does neointimal formation after arterial injury tell us about the critical mechanisms underlying arterial pathology in aging, atherosclerosis, or restenosis?

    Pharmacology of Neointimal Formation

    The best-studied model of neointimal formation is the response of the rat carotid artery to balloon angioplasty.23263132 This simplicity of the rat model has facilitated kinetic analyses of the smooth muscle response, leading to the definition of four waves describing the smooth muscle cell response to injury and to the identification of molecules playing a role in, or interacting with, each of these waves in rats.213334

    The use of the rat arterial response to injury as a model for human atherosclerosis or restenosis, however, should only be considered with caution. First, unlike arteries in larger mammals, the rat carotid artery has only rare intimal cells.3536 Formation of new intima may poorly replicate the response to injury when an intima or even an atherosclerotic lesion already exists.37 Second, although platelet adhesion occurs in injured rat arteries, thrombus formation and leukocyte infiltration are minimal. These important events are prominent after vascular injury in rabbits, swine, and nonhuman primates.38394041 In pig coronary arteries, injury caused by distension with an oversized stent initially causes thrombus formation, followed within a few days by significant macrophage and lymphocyte infiltration and finally by smooth muscle cell migration and proliferation in the intima.42 Leukocyte infiltration is also common in rabbit arterial lesions4344 but not in rat lesions.40 Unfortunately, little is known about the molecules controlling response to injury in these more complex systems.

    In the rat, the balloon injury model begins with complete destruction of the endothelium as well as extensive death of medial smooth muscle cells.32 The first response to balloon injury, called “first wave,” consists of medial smooth muscle cell proliferation and begins ≈24 hours after the injury. In elegant studies, Reidy and colleagues4546 have shown that this wave of replication can be completely accounted for by release of bFGF from dying smooth muscle cells. Despite extensive studies of PDGF as an in vitro mitogen,47 studies with infused PDGF, as well as studies with anti-PDGF antibodies, have shown that this molecule is only a weak mitogen for smooth muscle cells in the media of injured arteries.484950 However, more limited data suggest that α-adrenergic antagonists and Ang II may also control medial replication.515253

    Although the first wave has no obvious relation to the later migration and intimal proliferation, several studies with antisense agents directed at cell-cycle genes have shown that inhibition of these initial proliferative events in some as-yet-undefined way leads to a diminution of the final extent of neointimal proliferation. Since the antisense is likely only available in the first few days after injury, these studies suggest long-lasting effects on later events, including migration or intimal proliferation.54555657585960 The migration of smooth muscle cells across the internal elastic lamina to form the intima constitutes the “second wave.” Smooth muscle cells are readily observed on the luminal side of the internal elastic lamina 4 days after injury to rat arteries.32 The duration of migration is not known. Several molecules other than PDGF may contribute to smooth muscle migration, including TGF-β, bFGF, and, as discussed below, Ang II. The relative contributions of these different molecules are not known, nor do we know whether other molecules are involved.465361 PDGF has received attention because of many studies implicating this molecule as a growth factor for cultured smooth muscle.47 The main effect of PDGF in vivo, however, is on smooth muscle cell migration and not proliferation.495062 Immunoneutralization of endogenous PDGF or depletion of circulating platelets inhibits the development of the intimal lesion in response to balloon injury by inhibiting smooth muscle cell migration toward the intima without a reduction in the frequency of proliferating smooth muscle cells in the media or the intima. In the gentle denudation model, administration of PDGF-BB, which recognizes all isoforms of PDGF receptors, causes a 10- to 20-fold increase in smooth muscle cell migration but no more than a 2-fold increase in smooth muscle cell proliferation.48 In vivo dose-response studies show that discrepancies between these in vivo data and the potent mitogenic effects of PDGF on smooth muscle cells in vitro do not reflect inadequate dosage.48 As discussed below, we do not know whether PDGF is a mitogen in species other than the rat, as is suggested by studies of PDGF transfected into swine arteries. For that matter, we do not know whether PDGF is mitogenic in the rat at other sites of vascular injury, as is suggested by studies of PDGF infused into the kidney.63

    Once smooth muscle cells arrive in the intima of the rat artery, neointimal cells closest to the lumen may replicate for weeks to months.64 This replication is called the “third wave.” Although we cannot say that any specific molecule has definitively been identified as a third-wave mitogen, a few candidates appear to be present in the intima. For example, PDGF-A chain is overexpressed in the intima and colocalizes with replicating cells. Moreover, when PDGF-B was transfected into swine arteries, there was a marked elevation of replication.65 Unfortunately, we do not know whether this PDGF was acting as a growth factor in the usual sense or was mimicking the transforming mechanism of the viral homologue of PDGF, v-sis.666768 As already noted, in the rat model PDGF-B will not stimulate replication if infused, nor do antibodies to PDGF suppress third-wave replication.495069 Other growth control molecules that appear to be overexpressed in the rat neointima include the Ang II receptor, AT-1, and TGF-β.6170 IGF-1 is also overexpressed after injury; however, it is overexpressed in the media.71 The neointima can be stimulated to show a further increase of replication by infusion of several of these molecules. This increased responsiveness to mitogens can be called a “fourth wave” and involves at least TGF-β, bFGF, and Ang II as agonists.455161 Again, PDGF does not appear to be mitogenic in the rat carotid artery model, even at doses that markedly stimulate migration of smooth muscle cells in the second wave.48

    As the time this review was written, no specific molecular antagonist had been shown to inhibit this replication. Even antibodies to bFGF, which are so effective in inhibiting the first wave, have been shown to be impotent against third-wave replication.46 In the absence of inhibitors, Koch’s postulate cannot be fulfilled. Therefore, we cannot identify the critical molecules that sustain elevated replication in the third wave.

    Although we have emphasized smooth muscle cell mitogens, pathological loss of smooth muscle quiescence could also represent the loss of growth inhibitors. Regeneration of the endothelium inhibits proliferation of the underlying neointimal cells.326472 At this time, however, the mechanisms for this inhibition remain incompletely understood. Suppression of neointimal cell growth might be due, for example, to endothelial production of NO. In vitro studies have shown that NO can inhibit replication of cultured smooth muscle cells.73 Administration of large doses of l-arginine aimed at chronically raising NO levels in animals immediately after balloon catheter–induced endothelial denudation have resulted in significant inhibition of neointimal thickening.747576 Interestingly, Farhy et al77 have reported that L-NMMA, a blocker of NO generation, can partially counteract the suppressive effect of ramipril, an ACE inhibitor, on neointimal thickening after balloon injury in rat arteries.77 They speculate that inhibition of neointimal formation by ACE inhibitors may be a result of elevation in the level of bradykinin. Bradykinin is a potent stimulator of NO production and is itself normally degraded by ACE. When administered alone, however, the same dose of L-NMMA did not affect neointimal thickening.77 Moreover, Hansson et al78 have reported that the cytokine-inducible isoform of NO synthase is chronically expressed at high levels by smooth muscle cells at the denuded surface of the neointimal lesion in the rat injured carotid artery,78 an area in which smooth muscle DNA synthesis remains chronically elevated.64 In addition, in vitro studies suggest that in the presence of bFGF, NO can actually stimulate DNA synthesis and mitogenesis in smooth muscle cells from the rat aorta in primary culture, whereas a similar treatment causes inhibition of DNA synthesis in the passaged smooth muscle cells.79 In summary, we do not have definitive evidence for an endogenous role of NO as an inhibitor of replication of neointimal smooth muscle. Table 2 summarizes the molecules that control the three waves.

    Potential Clinical Targets Based on the Rat Model

    Angiotensin is of special interest as a mediator of neointimal formation because ACE inhibitors that block neointimal formation in the rat model5380 failed to inhibit restenosis in humans.81 Exploring the reasons for the failure of clinical trials using an ACE inhibitor requires a brief review of some of the pharmacology of the angiotensin system. The ultimate precursor of Ang II is the plasma protein angiotensinogen, from which renin, a kidney-derived endopeptidase, releases the inactive decapeptide Ang I. The carboxypeptidase ACE then metabolizes Ang I to release active Ang II. As already noted, ACE also contributes to the degradation of bradykinin, a potent endogenous inhibitor of neointimal formation.77 Although enzymes other than ACE appear to be able to activate Ang I,82 the lack of specificity of ACE inhibitors can be overcome by use of specific inhibitors directed at the angiotensin receptors.8384

    Components of the angiotensin pathway may be found together in the intima or neointima.8586 Although both AT-1 and AT-2 receptors are also present in the normal wall, the principal Ang II receptor, AT-1, is elevated in the neointima.7087 Angiotensinogen, which is available from the circulation, may also be produced by the vessel wall. Rakugi et al88 have reported an increase in angiotensinogen gene expression in both the media and the neointima during the first 2 weeks after balloon injury to the rat abdominal aorta. Although the validity of these data may be questioned in view of contradictory in situ hybridization studies from other authors,8990 angiotensinogen mRNA is available locally from perivascular fat. The vessel wall may also contain reninlike activity.91 Some of this activity may be chymase, a protease found in human mast cells, especially around blood vessels.82 ACE itself is made by the medial smooth muscle cells and by the endothelium, the major source of this enzyme.92 Rakugi et al88 have reported that despite loss of the endothelium, 2 weeks after balloon injury the rat abdominal aorta expresses increased levels of ACE activity as well as increased immunoreactivity for ACE in the neointima. Fishel et al93 also have reported increased ACE activity in the rat injured carotid artery as well as a tissue gradient of ACE expression, with the greatest immunoreactivity in the most luminal cells of the neointima. Upregulation of ACE activity also occurs in dog arteries 30 days after balloon injury.94 On the other hand, Viswanathan and colleagues7087 used quantitative autoradiography with iodinated ligands to study ACE expression up to 30 days after injury in the rat carotid artery and thoracic aorta. In these studies, levels of binding to ACE levels per milligram of tissue were similar in neointimal lesions devoid of endothelium 15 or 30 days after injury compared with sham arteries with endothelium. Thus, after balloon denudation, there may be no loss of ACE activity, since neointimal ACE replaces the ACE normally produced by the endothelium.

    In summary, a possible role for the renin-angiotensin system is supported by evidence for the expression of ACE and other components of the angiotensin system in the injured vessel wall. First-wave replication, migration into the intima, and intimal thickening can be prevented by angiotensin receptor agonists directed against the AT-1 receptor.8384 ACE inhibitors and receptor antagonists seem to block migration as well.8384 These effects are likely mediated by the AT-1 receptor. One report of inhibition by an AT-2 receptor ligand molecule required that the drug be given locally at very high doses.95 Thus, it is quite reasonable to interpret the effects of ACE inhibitors in blocking neointimal formation as being in large part the result of blocking formation of Ang II.80

    The clinical trials may have failed for several reasons. First, the effects of ACE inhibitors may be determined by genetic variation in the population. Ohishi et al96 recently found a striking correlation between restenosis and an isoform of ACE. This isoform, a deletion, had previous been identified by Cambien et al97 as a risk factor for myocardial infarction. Second, it is important to note that ACE inhibitor doses used in the clinical trials were ineffective in the rat studies as well as in trials after arterial injury in swine and nonhuman primates.9899100 Effective doses in animal studies were hypotensive,80 possibly because of the elevated levels of bradykinin discussed above. Such doses would not be acceptable in humans. Third, as already noted, ACE is not the only enzyme able to generate Ang II in humans. Chymase is present in human plaques and cannot be inhibited by the converting enzyme inhibitors.82101 Chymase activity is also dramatically upregulated 30 days after balloon injury in dog arteries.94 Other potential alternative pathways for the activation of Ang II also include serine proteases, such as leukocyte-derived tonin,102 and carboxypeptidases from human platelets103 or from the tunica media.104

    The PDGFs are also of special clinical interest because of the emphasis on these molecules in the “response to injury” hypothesis.47 We have already noted that in contrast to FGF or Ang II, infused PDGF-BB seems to function primarily in promoting migration.484950 The absence of a mitogenic effect in the rat model, of course, does not rule out an effect in humans. As already discussed, transfected PDGF-BB was mitogenic in swine. Moreover, again in the rat, the production of PDGF-B by cultured smooth muscle cells does correlate with an enhanced ability to grow without serum-derived growth factors.105106 We have recently found that ≈10% of the most superficial cells in the neointima express PDGF-B chain, as demonstrated by in situ hybridization.107 It is intriguing to think that the subset B chain–positive cells in vitro are the ancestors of the characteristic B chain–positive cells seen after passage in vitro. We do not know whether similar cells might exist in proliferative regions of human restenotic tissue.

    PDGF-A chain is also of interest, despite the observation that PDGF-A is itself a nonmitogen or a weak mitogen in vitro.48 This molecule is chronically overexpressed by smooth muscle cells in the neointima in the rat model69 and in atherosclerotic plaques in humans.108109 A recent immunocytochemical study colocalized PCNA, a marker for cell replication (see below), with PDGF-A chain.110 The mitogenic effect of a number of mild mitogens, including bFGF, TGF-β, and Ang II, has been shown to be substantially blocked in vitro by antibodies to PDGF-A.49 Thus, we need to consider the possibility that the localized overexpression of PDGF-A seen in the neointima may act as a cofactor that increases the fourth-wave responsiveness to other growth factors.

    TGF-β may also play an important role in third- and fourth-wave replication. As already noted, there is an accumulation of TGF-β in the neointima,61 and we found that infused TGF-β was a neointimal mitogen. More recently, Nabel et al111 found augmented neointimal hyperplasia in a swine model after injury and transfection with a TGF-β plasmid. Similarly, Wolf et al112 found that neointimal formation in the rat model was inhibited by antibodies to TGF-β. Finally, a recent study of atherectomy specimens found elevated levels of TGF-β in restenotic lesions.113

    These in vivo data contrast with the recent proposal by Grainger and colleagues114 that TGF-β in humans is an inhibitor of atherosclerosis. The Grainger hypothesis is based on studies in vitro in which TGF-β is sometimes seen as a growth stimulant and at other times as an inhibitor. This variability may reflect the strain of smooth muscle cells studied and their state of confluence.115116117 Grainger et al114 emphasize the inhibitory effects and attempt to link endogenously produced TGF-β with elevated atherosclerosis risk due to elevated Lp(a). Although Lp(a) is well known as a risk factor for atherosclerosis, a recent study suggests that Lp(a) levels also correlate with an increased incidence of restenosis.118119 As a homologue of plasminogen, the apoprotein of Lp(a), called Apo(a), is believed to inhibit the formation of plasmin.120121 Plasmin, in addition to its role in fibrinolysis, is essential to the activation of TGF-β.122 Grainger and colleagues114123124125 found that Apo(a) is mitogenic for smooth muscle cells in culture and were able to attribute this mitogenesis to inhibition of the formation of plasmin and the subsequent inability of the cells to activate TGF-β. In this view, the predominant role of TGF-β would be as an endogenous inhibitor of lesion formation at sites of active coagulation, and Apo(a) would enhance lesion formation by diminishing the production of activated TGF-β.

    Thrombin is another neointimal molecule that is attracting renewed interest because of the recent cloning of a thrombin receptor and the recognition that thrombin can have a direct mitogenic effect on cells independent of the coagulation cascade.126127 Particularly intriguing is the evidence that thrombin activity is chronically elevated in the neointima and that the neointima, as well as human atherosclerotic plaque, overexpresses the thrombin receptor.128129130 In addition, studies involving animal model systems more complex than the rat, especially the pig coronary artery stent model, suggest that thrombosis may well play a key role in the early events leading to restenosis.42

    The last three molecules for this discussion are somatostatin, heparin, and the integrins associated with thrombosis and vascular responses to injury. Somatostatin analogues and heparin-like compounds are of special interest because of the extensive studies of their ability to inhibit neointimal formation in animal models. Somatostatin analogues, in particular, angiopeptin, have been found to be effective in a range of animals species and different models of arterial injury, including fat feeding, balloon angioplasty, neointimal formation in vein grafts, and transplant atherosclerosis.131132133134135 Clinical trials are under way, but preliminary results appear to be equivocal.136 The pharmacological rationale for this approach is unclear; however, it is important to note that somatostatin receptors are widespread in the body and may act as vasodilators or vasoconstrictors in blood vessels.137138139140 Failure or success of angiopeptin trials to prevent restenosis will be especially important, because the dosages of the drug that were used in humans were comparable to those used in animals.

    Similarly, heparin has been widely studied as an inhibitor of intimal formation in animal models.141 As with somatostatin, the mechanism of action of heparin as an inhibitor of intimal hyperplasia is poorly understood. Although much attention has been focused on the potential role of heparin on c-myb, this molecule is expressed in the late G1 phase of the cell cycle and likely represents only one of several defects when growth is inhibited.142 Equally intriguing are the role of heparin in inhibiting migration and suggestions from Lindner and Reidy45 that a major action of heparin may be to wash bFGF out of the injured vessel wall after the mitogen is released from dying cells. Unfortunately, clinical trials using heparin to prevent restenosis have been disappointing.143144 Ellis et al143 administered intravenous heparin to patients over the first 18 to 24 hours after angioplasty and found no difference in the restenosis rates of patients treated with heparin compared with those given a dextrose infusion (41% versus 37%, respectively; P=NS). Similarly, a subsequent attempt to limit restenosis with a single daily subcutaneous injection of 10 000 IU heparin was halted prematurely because of the higher rates of restenosis and clinical events in the heparin treatment group compared with the usual care group.144 This lack of clinical benefit is difficult to explain but may be related to a rebound coagulation phenomenon associated with interrupted anticoagulant treatments.145 Edelman and Karnovsky146 suggest that differences in heparin dose scheduling may be critical. For example, the antiproliferative effect of heparin requires that the drug be administered for at least 4 to 7 days after injury in the rat carotid balloon injury model.32 Furthermore, cell proliferation and the ratio of the intimal to medial area are made worse when rats are treated with heparin dosages and administration schedules similar to those used clinically.146 Low-molecular-weight heparins also appear to be ineffective for the prevention of restenosis.147

    The failure of antiproliferative therapies based on angiotensin, somatostatin, and heparin may be telling us that we should consider other mechanisms as being critical to restenosis. A clue as to such mechanisms may have arisen serendipitously from a recent clinical trial directed at the thrombotic events occurring immediately after angioplasty. As part of the EPIC study, the chimeric monoclonal antibody Fab fragment (c7E3), which is directed against the platelet glycoprotein IIb/IIIa receptor, was administered to patients undergoing angioplasty or atherectomy who were at high risk for ischemic complications.148149150 This therapy resulted in a reduction in acute ischemic complications (eg, nonfatal myocardial infarction and emergency revascularization procedures), although at the risk of increased bleeding complications. Although it is easy to imagine that these trials are affecting only platelets, these antagonists to glycoprotein IIb/ IIIa, a platelet adhesive protein, may also affect a closely related integrin, αvβ3. Antagonists to αvβ3 have recently been shown to block smooth muscle migration in vitro and intimal formation in vivo.151 Moreover, we have found that this same receptor is required for movement of smooth muscle cells in response to osteopontin, an abundant and specific marker of intimal smooth muscle cells in the rat152 and in human plaque.153154 Therapies targeted against integrin receptors involved in platelet aggregation and cell migration represent an intriguing new direction.

    Role of Intima in the Formation of an Atherosclerotic Lesion

    Stary et al155 note that the initial lipid accumulation occurs deep in the wall, within the preexisting intimal mass. The intimal mass is not defined as atherosclerosis, since it has not yet accumulated lipid and is seen as a normal feature of vessels in our species. Aside from such semantic issues, localization and initiation of atherosclerosis in intimal masses are reminiscent of experiments in balloon-injured animals in which lesions develop selectively at sites of previous injury and intimal formation.37 Thus, atherosclerosis may begin in the “soil” of the intima. The peculiar properties of the intima that lead to such a phenomenon have not been identified. However, there have been suggestions that local accumulations of specific glycosaminoglycans related to chondroitin sulfate may promote local lipid accumulation.156157

    The time course of intimal smooth muscle proliferation in relation to lipid accumulation in humans is not known. Ross47158 has suggested that proliferation of smooth muscle cells is important in advanced lesions. This idea, however, is not supported by cell kinetic studies measuring either the frequency of cells able to incorporate labeled thymidine ex vivo or the number of cells identified as replicative on the basis of staining for a cell cycle–specific marker.159160 The low replicative rates seen in advanced lesions correlate with studies of cells cultured from lesions. Moss and Benditt161 were the first to culture these cells. They found that plaque smooth muscle cells have a greatly shortened life span relative to normal medial cells. This observation has been reproduced by others.162163 More recently, we have extended this observation by looking at apoptosis. Plaque smooth muscle cells, at least in vitro, have a very high rate of spontaneous cell death. This high level of apoptosis may account for the apparent short replicative life span and suggests that plaque cells have undergone some as-yet-undefined injury in vivo.164

    Interestingly, the only actual evidence that smooth muscle proliferation occurs in atherosclerosis depends on evidence that the lesions are monoclonal or oligoclonal. The original observation of clonality by Benditt and Benditt5 has been reproduced by two other groups.234 It is difficult to imagine how clonality could arise unless proliferation plays a critical role in an event at some time in the ontogeny of the lesions.

    Hansson et al165 have recently shown that lymphocytes in lesions are polyclonal. The remaining cells in these lesions are macrophage, smooth muscle, and endothelial cells. Although plaque macrophage and endothelial cells do replicate,159166 there are no known examples of these cell types forming monoclonal growths other than in neoplasms.47159 Thus, monoclonality implies that smooth muscle proliferation must occur during the formation of the lesion and that the initial group of cells giving rise to the lesion must be very small. Such an early expansion of intimal smooth muscle mass has been described exactly to occur in the proximal left anterior descending coronary artery, a common site for occlusive coronary artery lesions in adults.1417 If the increase in mass described in the newborn left anterior descending coronary artery correlates with smooth muscle replication, then very few further doublings may be required to account for the mass of smooth muscle seen in an adult lesion.

    In summary, the available direct evidence suggests that smooth muscle replication occurs very early in the formation of atherosclerotic lesions.1417 We cannot rule out the possibility that smooth muscle replication occurs at a very low rate over several years or that replication occurs at a high rate in an episodic fashion.

    Special Properties of Atherosclerotic Intima

    Whether or not the atherosclerotic plaque is hyperproliferative, the lesions are progressive, and it seems obvious that progression must depend on unique properties either of the intima or of the plaque itself.167 Table 1 is an attempt to collate those properties with special regard to genes that are underexpressed or overexpressed by the intimal or plaque smooth muscle cell relative to medial smooth muscle cells. This table emphasizes rat and human data, the species about which the most is known. Some rabbit data are also included. In addition, the table includes data from cultured smooth muscle cells, since the preservation of properties in vitro identifies intimal smooth muscle cells that may be differently differentiated from medial smooth muscle cells.

    Table 1 also lists a large number of molecules whose expression is characteristically decreased by intimal cells compared with medial cells. Loss of expression of the genes associated with the fully differentiated, “contractile” phenotype was first described by Campbell and Campbell168 and has been confirmed by others.169170171 Campbell and Campbell have given the change from a contractile to a “synthetic” phenotype the name “modulation.” The term synthetic is used because cultured cells lose much of their microfilaments and, as seen by transmission electron microscopy, acquire an extensive rough endoplasmic reticulum. Because modulation coincides with the onset of replication in vitro, several workers have proposed that special properties of the modulated smooth muscle cell may include proliferation and synthesis of matrix molecules that contribute to formation of the intima and the plaque.168169170171 Molecules lost in modulated cells include smooth muscle myosin, desmin, caldesmon, and α-actin.10172

    Unfortunately, the term synthetic has been loosely applied to cells lacking a smooth muscle phenotype, because intimal cells as well as many cells in the media may lack the contractile phenotype without showing evidence of an active synthetic apparatus169173 (D. Gordon and S.M. Schwartz, unpublished data involving the internal mammary artery, 1994). Inversely, smooth muscle cells can rapidly leave quiescence in the vascular wall after balloon injury, enter S phase, and still express abundant α-actin mRNA.174 Thus, the equation of synthetic phenotype with replication is unconvincing. Similarly, the media of avian aortas contains a second cell type that, while apparently quiescent, lacks morphological features of the contractile smooth muscle cell.175176177 Thus, loss of the contractile phenotype should be distinguished from the onset of proliferation or change to a phenotype associated with high levels of protein synthesis.

    An excellent example of the difference between a vessel with only a media and one with an atherosclerotic intima is the issue of vascular contractility. Normally, the vessel wall exists in a relaxed state due to the endogenous production of NO by endothelium. NO-dependent relaxation, however, is greatly impaired in atherosclerotic vessels.178 This loss of function is not due to a change in the smooth muscle of the media. Loss of NO function has been attributed to endothelial injury or to the inactivation of NO by free radicals produced in the plaque.179 In contrast, Joly et al180 described the appearance of the inducible form of NO synthase that follows balloon injury. Similarly, as described above, the neointima overexpresses angiotensin receptors, and the plaque has been shown to overexpress endothelin.181 Although we have not yet distinguished the effects of atherosclerosis from those of the more general appearance of the neointima, our point is simply that we might expect the vasomotor activity of a diseased artery with an intima to be very different from vasomotor regulation in a normal artery. The contribution of this altered pharmacology to the ability of the vessel wall to maintain a normal lumen caliber is largely unexplored.

    In summary, Table 1 points out that intima and media are quite different tissues. At least some genes are overexpressed in the intima, even in the absence of atherosclerosis; for example, the human intima overexpresses the β1 chain of laminin and the ED-A splice form of fibronectin.182183 In the rat, a large number of genes are overexpressed after injury, but so far only tenascin has been shown to be overexpressed in the few intimal cells seen in the normal rat artery.184185 Tenascin, β1 laminin, and ED fibronectin are important, since their expression suggests that intimal smooth muscle cells rather than merely lacking the properties of medial smooth muscle may have their own unique properties. We have already discussed the possible significance of overexpression of NO synthase, AT-1 angiotensin receptors, proteoglycans, PDGF, or TGF-β in the ontogeny of smooth muscle proliferation and lipid accumulation. Differences in NO metabolism186 or AT-1 receptors,87 to cite just two markers, are likely to mean that vessels with an intima have very different pharmacology than vessels with only a media. A critical issue, as we will see below, is whether the unique properties of intimal cells represent a unique cell type with heritable properties that distinguish intimal smooth muscle from medial smooth muscle.

    Intimal Cells Overexpress Certain Genes In Vitro as Well as In Vivo

    Cell culture studies suggest that intimal cells may be differentiated from the typical medial smooth muscle cell. Smooth muscle cell lines cultured from 2-week-old rat pups have several unique properties. These include an epithelioid morphology, the ability to grow without platelet-released growth factors, and secretion of PDGF.187188 These same properties appear when neointimal cells, isolated 2 weeks after arterial injury, are placed in culture.105 When the same cells were studied for gene expression, a group of genes was found that shows overexpression or even unique expression in pup and intimal cells compared with normal medial cells cultured from adult arteries.187189190 This collection of properties and genes led us to suggest that the vessel wall, at least in the rat, contains two types of cells: π and μ cells. Genes overexpressed by π cells include PDGF-B chain, CYPIA1, elastin, and osteopontin.187189190

    Unlike the distinction between contractile and synthetic phenotypes, which is lost when all cells put in culture become synthetic, the differences between the μ phenotype and π phenotype seem to be maintained in passage, and cells with π or μ properties can be isolated by cloning cells from mixed cultures.191192 Moreover, some of these genes, although first identified in vitro, are also overexpressed or uniquely expressed in the neointima in vivo. At least for the rat, we suggest that smooth muscle cells contain two distinct “cell types,” one of which takes a special part in the formation of the neointima.

    Comparable evidence has not yet been discovered for subsets in human smooth muscle. Cultured human fetal smooth muscle cells also differ from adult smooth muscle cells in their expression of two homeobox genes, HoxB7 and HoxC9. These genes are also seen in pup cells but not in rat cells, however. HoxB7 and HoxC9 are not seen in human intima or atherosclerotic plaques (J.M. Miano, A.B. Firulli, E.N. Olson, P. Hara, C.M. Giachelli, S.M. Schwartz, unpublished data, 1994). However, as already noted, human intima preserves certain genes that are also seen in the fetal vessel.10 Moreover, if atherosclerotic lesions are monoclonal, the origin of lesions in the intima suggests that lesions are derived from a unique subset of vessel wall smooth muscle cells localized to this layer.5

    Mechanisms Controlling Plaque-Specific Gene Expression by Smooth Muscle Cells

    Overexpression of certain molecules by intimal cells might be the result of two quite different, but not necessarily independent, mechanisms. First, as suggested above, intimal cells might belong to a unique lineage either derived from a common precursor or differentiated from typical smooth muscle cells. The detection of stable π and μ phenotypes in vitro supports this hypothesis. Alternatively, and more obviously, plaque cells may show overexpression or repression of certain molecules as a result of mediators present in the plaque environment. Particularly important among such mediators are oxidation products or more traditional inflammatory mediators.193194 Collins195 has suggested that a common factor linking inflammation and oxidation is the role of nuclear factor-κB as a trans-acting factor induced by oxidized radicals and by many cytokines.195 Table 1 includes plaque-specific genes whose overexpression might be explained as an inflammatory response. In some cases, however, this may be misleading, since overexpression in the plaque is permanent, whereas overexpression in response to cytokines is often only transient. Distinguishing between extrinsic and endogenous controls of gene expression is likely to depend on cell culture results showing stable expression, as is evident for some genes in Table 1, as well as promoter analyses taking place in the individual labs.

    Pathological Significance of Plaque-Specific or Intimal-Specific Genes

    Among the more interesting genes shown to be overexpressed in plaques are tissue factor, MCSF, GMCSF, osteopontin, and bone morphogenic protein.

    Tissue Factor

    The normal arterial wall contains little or no tissue factor. The time course for the appearance of tissue factor in plaques is not known, however. Tissue factor is prominent in cells of the plaque, including smooth muscle cells. Davies and Thomas196 and others197198199200 have suggested that thrombosis is a critical event in plaque progression. Thus, it is likely that accumulation of tissue factor is critical to the morbidity of lesions. Moreover, Taubman201 has shown that tissue factor mRNA expression was elevated after balloon injury in rats. This was not surprising, since tissue factor was known from in vitro studies to be a component of the early phases of the cell cycle. Perhaps more surprisingly, he found an elevation of tissue factor activity, implying that a procoagulant response is part of the formation of neointima.

    MCSF/GMCSF

    The presence of leukocyte growth factors MCSF and GMCSF as well as receptors for leukocyte factors (see Table 1) is of special importance because of growing evidence that plaque macrophage, rather than plaque smooth muscle cells, may constitute the major unique proliferative element in the plaque.159160 The unique properties of the proliferative plaque macrophage have yet to be explored.

    Osteopontin and Bone Morphogenic Protein

    Osteopontin and bone morphogenetic protein 2a have both been found in smooth muscle cells of atherosclerotic plaques.154202203 The presence of these molecules is supportive of the hypothesis that plaques are derived from unique subsets of smooth muscle cells, because osteopontin was first found in neointimal cells and in the rat pup artery as part of the π phenotype.190 Bone morphogenetic protein 2a, on the other hand, is also seen only in plaques. Intriguingly, cells found to express bone morphogenetic protein 2a also express immunocytochemical markers associated with a special form of smooth muscle cell, the pericytes seen around small vessels, again suggesting a unique lineage for plaque smooth muscle compared with medial smooth muscle.203 The association of osteopontin and bone morphogenetic protein 2a with areas of calcification in the plaque also suggests that the mechanisms of bone mineralization may also play a role in vessel wall calcification.

    Mechanism of Lumen Occlusion in Atherosclerosis

    Vascular lumen size can be regulated either by rapid changes in smooth muscle tone or by structural rearrangement of cells and extracellular matrix components of the wall. The latter mechanisms include “remodeling” and “elastic recoil.” For example, Glagov et al204 noted that human vessels can undergo massive accumulations of atherosclerotic mass without lumen narrowing. The vessel compensates for the new mass by undergoing a compensatory redistribution of vessel wall mass so that lumen size is maintained. This remodeling permits a normal level of blood flow until an adaptational limit is exceeded. This limit appears to occur when ≈40% or more of the area bounded by the internal elastic lamina is occupied by intimal mass. Decreased lumen size in atherosclerosis may depend on pharmacological mechanisms associated with the neointima or the plaque that cause a failure of normal remodeling events. In contrast, elastic recoil, an expression often used to describe the rapid loss of lumen gain after angioplasty, describes the resistance of the vessel wall component to the mechanical stretch imposed by the inflated balloon catheter.

    Remodeling is a normal response that allows the vessel to maintain normal levels of blood flow and wall stress and can be seen in small muscular arteries as well as in large elastic arteries.205206 Branching patterns of conduit vessels seem to be genetically determined.207 Thus, the only way arteries can respond to a demand for an increase or a decrease in blood flow is by changing vessel caliber. These changes, in some cases, occur by rearrangement of existing vessel wall components rather than synthesis of new mass or cell replication.208

    Despite the observations of Glagov et al204 of arterial remodeling during atherogenesis in human coronary arteries (above), remodeling has been largely ignored in animal models of arterial injury and repair. The extent of remodeling versus loss of lumen due to intimal formation is illustrated by an important experiment by Jamal et al.206 When Jamal et al balloon-injured the rabbit carotid artery, they found the expected neointimal hyperplasia. However, the animals showed no narrowing of the lumen despite a 100% increase in total wall thickness attributable to the neointima. In contrast, the same study showed a significant (14%) narrowing in response to an experimental restriction of flow in the vicinity of the thyroid artery where endothelium had regenerated. This effect of the endothelium in an injured vessel is consistent with observations that structural adaptation to changes in flow requires an endothelium.206 Since reendothelialization generally correlates with a diminution of intimal thickening, these data may even imply a negative correlation of intimal mass with luminal narrowing.72

    The role of the endothelium in controlling lumen size may be especially important given the often neglected fact that plaques have a prominent microcirculation that comprises capillaries arising from the adventitia.209210211 The role of these small vessels in regulating structural change or, for that matter, contractile properties of the atherosclerotic vessel remains unexplored.

    If the correlation of intimal (or plaque) mass with lumen caliber is not a simple one, how do we account for angiographic changes seen after aggressive lipid-lowering therapy?212213214 It is important to realize that the reported clinical benefit of these studies far exceeds the improvement of stenosis diameter. The improvement in lumen size in these studies is very small (eg, 0.7% to 5.3%, or an increase of 0.003 to 0.117 mm in minimum absolute diameter).212214 To put these results in perspective, one should note that 6 months after percutaneous coronary angioplasty, there is an average improvement of 16% diameter stenosis units and a 0.47-mm increase in minimum absolute diameter.215 Moreover, even these modest changes in lumen diameter could reflect changes in adherent thrombotic material or state of vasospasm rather than a decrease in mass of the atherosclerotic intima.216 Rather than opening the lumen, lipid-lowering therapies may improve clinical outcome by stabilizing the lesion.217 Davies and colleagues196218219 and others199200 have suggested that the formation of fissures in plaques is the critical step leading to vascular occlusion. Davies’ hypothesis would imply that we may be able to develop diagnostic tests, based on plaque composition as assessed by magnetic resonance imaging, IVUS, or even gene expression patterns in atherectomy specimens, that might indicate lesion prognosis or the effectiveness of drugs targeted at stabilizing the lesion.

    These same tests may allow us to estimate changes in vessel wall mass much as the advent of roentgenography led to estimates of tumor cell growth. The ability to study the progress of human lesions may be critical to our understanding of how lesions progress and eventually kill. Animal models with advanced disease are rare, with the exception of the spontaneously hypercholesterolemic swine.220 Autopsy data in humans are also confusing. Serial angiographic studies in humans suggest that the majority of myocardial infarctions may occur because of thrombotic occlusion of arteries that previously did not contain hemodynamically significant stenoses (eg, <50%).221222 However, postmortem studies do not bear this out. For example, Qiao and colleagues223224 have reported that in both native coronary arteries and saphenous venous bypass grafts, atherosclerotic plaque rupture with thrombosis most commonly occurred at sites with severe narrowing (eg, >90% area stenosis).

    The Nature of Restenosis Following Angioplasty

    Fig 2 shows the tissue that characterizes coronary artery restenosis as seen in atherectomy specimens or at autopsy. Despite use of the word “proliferative” by pathologists to describe this tissue,225 as we have already suggested, it may be entirely incorrect to assume that the formation of a neointima in a balloon-injured rat carotid artery is a model for the loss of arterial caliber seen after angioplasty, ie, “restenosis.”

    We have used immunocytochemical labeling for the PCNA159226 to determine the proliferative profile of 100 restenotic coronary atherectomy specimens.227 To our surprise, the vast majority of the restenotic specimens (74%) had no evidence of PCNA labeling. Moreover, in those specimens with proliferation, only rare PCNA-positive cells were present, and there were no differences in the frequency of PCNA-positive cells in restenotic specimens collected in the first 3 months, 4 to 6 months, 7 to 9 months, or >9 months after the initial interventional procedure. Furthermore, only 12 of 30 specimens obtained within 60 days of the initial coronary interventional procedure had one or more PCNA-positive nuclei per slide (including nine specimens collected within 6 days of the initial procedure, only three of which had immunolabeling of 1, 7, and 20 cells per slide). A similar study using in vitro bromodeoxyuridine labeling also found low levels of proliferation in restenotic atherectomy specimens,228 and Strauss et al229 found no PCNA-positive cells in atherectomy specimens from seven restenotic stented coronary artery lesions. In contrast to these studies of replication in restenotic coronary atherectomy tissue, Rekhter et al,230 using the same methodology, found PCNA immunolabeling of rapidly stenosing human arteriovenous hemodialysis arteriovenous fistulas to be high. These three studies of restenosis suggest that the response of human atherosclerotic coronary arteries to balloon angioplasty lacks the proliferative first wave seen in the rat model. If this is true, we may not expect clinical trials of drugs directed at the first wave to be successful.54

    Our results and those of Strauss et al229 contrast with a recent report by Pickering et al.231 All restenotic coronary and peripheral arterial specimens had surprisingly high percentages of cells that were considered PCNA-positive; eg, as high as 59% of cells were PCNA-positive, comparable to values in malignant neoplasms.232 Surprisingly, given the very high values, none of these specimens were obtained within 1 month of the initial interventional procedure. It is unlikely that mean PCNA labeling indices of 15% to 20% are physically possible in atherosclerotic coronary arteries, where small changes in vessel wall mass can result in dramatic changes in residual luminal diameter. Although we assume that there is some error in the data of Pickering et al, the detection of differences in the frequency of PCNA-positive cells in their study nonetheless raises concerns that our method might be insensitive to low levels of replication. These cell kinetic studies suggest that we reevaluate pathologists’ use of the term proliferative in describing Fig 2. For example, Nobuyoshi et al225 examined 39 dilated lesions from the postmortem coronary arteries of 20 patients who had undergone angioplasty. The extent of intimal proliferation was defined by the histological appearance of stellate fibroblast-like cells with a myxomatous appearance. It is essential to note that the term proliferative as used here is a morphological term, not a measure of replication, such as thymidine index, mitotic frequency, or even the PCNA values used in our studies. Cells having this proliferative morphology are not unique to restenosis. Similar cells are commonly seen beneath the endothelium in areas of nonatherosclerotic intimal thickening as well as in primary atherosclerotic coronary artery lesions that have never been exposed to an interventional device.233234 Although the myxomatous tissue seen in atherosclerotic or restenotic lesions does not appear to be actively proliferative, most observers agree that there is an increase in the amount of this tissue in restenotic versus primary lesions.159227233234235236

    We do not know whether the increase in amount of proliferative tissue represents a redistribution of the components of lesions due to compression by the catheter or some reaction such as formation of extracellular matrix, migration, or cell proliferation at a low level not measured by current methods. The appearance of proliferative tissue within the wires of stented coronary arteries supports the idea that this is not simply a redistribution of preexisting plaque components. To date, however, we lack direct histochemical evidence of replication.229 Perhaps the wires push into the wall, or the wall components migrate around the wires.

    Relevance of Animal Stenosis to Human Restenosis

    These data on replication force us to reconsider the relevance of animal models to the human problem. The issue is illustrated in Fig 3. Almost all animal studies, particularly those done in the rat, measure a decrease in lumen size from the initially normal situation. This can rightly be called stenosis. In contrast, the clinical problem is defined by “loss of gain,” which is the extent of dilatation lost after an atherosclerotic vessel has been dilated to achieve what the interventional cardiologist believes is an optimal diameter (eg, a popular clinical definition of restenosis is >50% loss of initial luminal diameter gain). This loss of gain, rather than the preexisting vessel caliber, is rightly called restenosis. By current angiographic criteria, even a return of the human vessel to its predilation diameter would be defined as restenosis, despite no change in intimal mass.

    Thus, stenosis, as seen in most animal models of arterial injury, is very different from the loss of gain called restenosis. Studies that equate intimal hyperplasia with restenosis may be especially misleading. For example, luminal narrowing after injury to the rat carotid artery is more pronounced after 2 weeks (75%) than after 12 weeks (35%), implying that early stenosis may be due to smooth muscle contraction of the vessel.24 Loss of lumen caliber could depend more on the extent of remodeling of the vessel wall to compensate for a change in mass than on the intimal mass itself.204

    Using these definitions, we would consider the response of the rat carotid artery as a loss of preangioplasty caliber, ie, a true stenosis with a decrease in lumen size. To the best of our knowledge, angiographic studies in humans do not show that angioplasty produces a similar acceleration of loss of the preexisting lumen. This, of course, would be difficult to demonstrate angiographically because the initial lesion usually already has a critical stenosis. Furthermore, animal models, perhaps by design, require that all manipulated arteries become narrowed, ie, stenotic, as shown in Fig 3. There is no animal model in which 50% or any substantial percentage of manipulated arteries remains dilated beyond their initial unmanipulated caliber, yet this 50% rate (or higher) is the success rate seen when atherosclerotic human arteries are dilated with an angioplasty balloon.

    Perhaps we need an animal model for successful angioplasty. Only recently have larger animal models been developed that take remodeling into account after angioplasty.237238239240 Remodeling of blood vessels, however, has been discussed at length in the hypertensive microvasculature and in the response of larger vessels to changes in blood flow.208241 Of particular interest, Langille and O’Donnell241 showed that the initial response of a carotid artery to reduced flow is active vasospasm. After 2 weeks, however, this active process is replaced by a fixed remodeling that cannot be reversed by vasorelaxants. Finally, as already discussed above, Langille, working with Jamal et al,206 found no loss of lumen despite a 100% increase in wall mass due to intimal thickening. Similarly, Kakuta et al238 found that the extent of intimal change did not correlate with the loss of caliber in balloon-injured atherosclerotic rabbits. They propose that the major determinant of restenosis, again, is remodeling. Similar results have been found in the swine by Post et al.242

    One possible way of addressing these issues in humans is to ask whether restenosis is the result of an increase in wall mass or the result of remodeling of the vessel wall to reestablish its preangioplasty caliber without a change in wall mass. Our ability to evaluate human vessels is changing because of the use of IVUS to image the affected wall. Previous data, based on histological and imaging studies, suggest that plaque compression, disruption with fracture, and dissection of the intima and media and stretching of the more normal portions of the media are involved in creating a bigger lumen.243244245246247248249250 However, newer concepts are emerging. For example, Losordo et al251 used IVUS to study 40 patients immediately before and after iliac artery angioplasty. The areas of the arterial wall, plaque, lumen, and neolumen resulting from the procedure were examined. Over 70% of the increase in luminal area immediately after angioplasty was contained within the plaque fracture (the so-called neolumen). Plaque cross-sectional area decreased by approximately one third, but total artery cross-sectional area increased only minimally (≈5%) with the dilatation. Thus, the major effects of angioplasty may be to redistribute the components of the wall. Conversely, loss of lumen, ie, a combination of elastic recoil and restenosis, might be due to healing of the fissure rather than formation of new mass.

    Preliminary intracoronary ultrasound studies suggest that increases in plaque area with restenosis following angioplasty are actually small (eg, 5% to 7%).252 The clinical significance of this small increase in plaque mass in an artery that is already severely diseased is unknown. The authors speculate that intimal hyperplasia may not be a dominant factor in the restenotic lesion and that, instead, remodeling may account for ≈60% of late lumen loss.253 This concept of “chronic recoil” is similar to the phenomenon of vascular remodeling or to the wound-healing model discussed above.

    In summary, the available data do not demonstrate that cell proliferation is a major component of coronary restenosis. Some apparent restenosis at late times may even be the result of recoil occurring over the first few hours after angioplasty, followed by relatively minor additional changes resulting from plaque progression on wound healing. This sort of early recoil is called elastic recoil. Alternatively, restenosis could be the result of a redistribution of wall mass, analogous to wall changes described in hypertensive microvessels and to changes seen in arteries of experimental animals following angioplasty as described by Langille206 and Kakuta et al238 (above). If this is true, antiproliferative approaches to therapy, even with the elegant use of molecular biology to inhibit growth, may be irrelevant.54142 It is intriguing to note that a recent study in the swine-stent model actually showed an increase in restenosis when an injured wall was irradiated to prevent cell proliferation.254 As ultrasound technology improves, it will be possible to test these hypotheses on the basis of serial measurements of changes in vessel wall mass. The kinetics of those changes may be useful in estimating the expected rate of cell replication in tissues undergoing restenosis.

    Future Directions

    The central theme of the present review has been a discussion of the intima, the unique properties of intimal smooth muscle cells, and the possible role the intima may play in atherosclerosis and restenosis. Although we have been pessimistic about simplistic equations of intimal proliferation with loss of lumen in atherosclerosis or restenosis, this should not in any way diminish the obvious fact that proliferation must occur to some extent and at some time during both disease processes. This proliferation forms the intima, and the pathology of both of these clinical problems depends on the special properties of the intima. In atherosclerosis, it is the intima that accumulates fat, becomes calcified, expresses tissue factor, and ultimately breaks down, leading to occlusive vascular disease. Restenosis, of course, would not be a problem if we did not need to treat atherosclerosis. Moreover, any mechanism likely to account for restenosis is probably going to depend on new tissue formed after injury. This new tissue is important and may be the result of replication, migration, or extracellular matrix accumulation. Furthermore, this new tissue may contribute to lumen narrowing by occupying space, causing tissue contraction or geometric remodeling of the vessel wall. Even if replication contributes only a small number of cells to remodeling, it is possible that changes in phenotype associated with replication are also critical to the remodeling process.

    At a basic science level, we need to know three things. First, we need to know much more about why the intima is different from the media. As we have discussed above, there are two possibilities. Intimal smooth muscle cells may belong to different lineages, just as different skeletal muscle cell phenotypes appear during differentiation of that cell type. Definitions of “lineage” are rapidly changing because of the identification of genes and 5′ sequences that determine expression of cell type–specific proteins. Such determination elements have not yet been identified for smooth muscle cells. Presumably, the identification of such elements will lead to a much better definition of smooth muscle at a molecular level and, therefore, to an understanding of why intimal cells lose the usual patterns of gene expression seen by classic medial smooth muscle cells. On the basis of our data showing two distinct lineages in rat smooth muscle, we would like to imagine that similar mechanisms may eventually distinguish human intimal cells from medial smooth muscle cells.

    The second basic science issue to be determined is the nature of the inflammatory process in atherosclerotic intima. Like any chronic inflammatory tissue, the intima of an atherosclerotic plaque is an extremely confusing place. Diagrams purporting to explain behavior of the intima on the basis of these mediators are of limited assistance in identifying therapeutic targets unless we can identify individual critical processes or molecules. It would be intriguing, however, to see if this complexity might not be simplified by identifying a critical inflammatory agent or eliciting antigen among the products of oxidation, lipid accumulation, necrosis, and coagulation. Such a critical molecule would offer an ideal therapeutic target to inhibit lesion progression. Oxidation products are especially interesting, given evidence that antioxidants, independent of lipid levels, can cause lesion regression or failure of progression.255 Apo(a) and related components of coagulation are also intriguing candidates, especially given the recent surprising observation that human Apo(a) transgenic mice develop lipid deposition and atherosclerotic lesions despite lack of evidence that this apoprotein interacts with lipid or alters serum lipids.256

    The third basic science issue to be determined is the relevance of animal models. The failure of animal models to predict outcomes of trials with heparin, converting enzyme inhibitors, angiopeptin, and, more recently, lovastatin is disturbing.81140143144257 On the one hand, we could conclude that the best-studied model, the rat, is irrelevant, but many of these drugs have also appeared to work in other species. Alternatively, it seems to us that investigators in this field need to reconsider the premises underlying the design of the animal experiments. For example, the best understood part of the rat model is the first wave of medial replication. Much less is known about the chronic proliferation seen once the intima is formed, ie, the third wave. We also know virtually nothing about ways the neointima might contribute to lumen loss other than simple changes in mass. For example, to our knowledge, there are no studies of flow-dependent remodeling in the presence or absence of an intima despite the dramatic changes in lumen caliber that result from changes in flow. For that matter, we lack any mechanistic or molecular mechanisms to explain the changes in lumen size or wall mass attributed to remodeling. In that respect, concepts developed in the field of skin wound healing may prove useful, particularly the notion of granulation tissue contraction and remodeling.

    Another critical issue is the lack of animal models with the typical features of advanced lesions. To our knowledge, the only animal model that consistently produces a lesion comparable to those seen in occlusive coronary artery disease is the spontaneously hyperlipidemic swine. These animals show extensive plaque necrosis, vascularization, rupture, and lumen narrowing at a very high frequency.220 Unfortunately, these animals are not generally available. More commonly, studies are performed by using animals with fatty streaks or fibrous caps, lesions usually not associated with occlusive coronary artery disease in humans.

    At a clinical level, we need to know a lot more about how the intima contributes to lesions. Presumably, there are some basic properties of the intima that give rise to monoclonal lesion growth and initiate accumulation at focal sites. Could the properties of intima in specific sites explain why the branches of the renal artery are free of lesions but lesions are detected on the unbranched surfaces of the aorta? The observation that intima is needed for closure of the ductus arteriosus raises another set of questions.18 Do special properties of the ductus arteriosus intima control closure of this vessel? Does the natural closure of the ductus mirror processes also seen in pathological narrowing of atherosclerotic arteries?

    Finally, theories of both atherosclerosis and restenosis simplistically assume that an increase in mass of the intima is the cause of loss of lumen caliber. We know that this is not simply true, but we lack a more comprehensive hypothesis. Terms like late loss, chronic recoil, or remodeling simply put names on poorly understood processes. Concepts of plaque enlargement by mural thrombosis or breakdown of plaque with critical fissuring offer a tantalizing prospect of molecular targets for therapy that again depend on our knowing why these events occur in the progressing plaque. One can guess where advances are likely to occur. Better, preferably noninvasive, imaging methods are needed to give us more precise definitions of the clinical problems. This, in turn, will hopefully lead to the development of better animal models. Already, IVUS is providing new insights into the composition and extent of human lesions and how they respond to various interventions (Fig 3). In turn, tissue from humans and from better animal models should help us learn how to control expression of the molecules that lead to plaque narrowing and ultimately death.

    Selected Abbreviations and Acronyms

    π cells = pup intimal cells
    μ cells = medial unmanipulated cells
    ACE = angiotensin-converting enzyme
    aLDL-R = acetylated low-density lipoprotein receptor
    Ang I, Ang II = angiotensin I and II, respectively
    Apo = apolipoprotein
    AT-1, AT-2 = Ang II receptors
    bFGF = basic fibroblast growth factor
    BMP = bone morphogenetic protein
    Col I(a1) = α1 procollagen I
    CYPIA1 = cytochrome P-450
    ED = extra domain
    FGF = fibroblast growth factor
    GMCSF = granulocyte macrophage colony–stimulating factor
    ICAM = intercellular adhesion molecule
    IGF = insulin-like growth factor
    IL = interleukin
    IVUS = intravascular ultrasound
    L-NMMA = NG-monomethyl-l-arginine
    Lp = lipoprotein
    LPL = lipoprotein lipase
    MCP = monocyte chemoattractant protein
    MCSF = macrophage colony–stimulating factor
    MHC = myosin heavy chain
    NO = nitric oxide
    PCNA = proliferating cell nuclear antigen
    PDGF = platelet-derived growth factor
    SM = smooth muscle
    SMC = SM cell
    SR = sarcoplasmic reticulum
    TGF = transforming growth factor
    TNF = tumor necrosis factor
    VCAM = vascular cell adhesion molecule

    
          Figure 1.

    Figure 1. Top left, Histology of intima vs media. Coronary artery is from an adult patient with idiopathic dilated cardiomyopathy. In the absence of atherosclerosis, there remains diffuse intimal hyperplasia. The intima is several cell layers thick and composed of smooth muscle cells and is separated from the distinct media by the internal elastic lamina (arrows) (hematoxylin and eosin, original magnification ×200). Top right, Advanced atherosclerosis of the left circumflex coronary artery. This lesion was treated by percutaneous balloon angioplasty 23 days before the patient’s death. Note the complex nature of the lesion with a distinct fibrous cap (FC), intimal hyperplasia (IHP), intramural hemorrhage (H), and a necrotic core (NC) composed of inflammatory cells and accumulated lipid (hematoxylin and eosin, original magnification ×100). Bottom left, Myxomatous tissue. Directional coronary atherectomy specimen from a restenotic lesion shows stellate-shaped smooth muscle cells. Smooth muscle cells with this appearance are often regarded as evidence of proliferation. Note the homogeneity of these cells and the absence of inflammatory and endothelial cells in this region (hematoxylin and eosin, original magnification ×200). Bottom right, Directional coronary atherectomy specimen from a primary lesion. Again, note the presence of stellate-shaped smooth muscle cells but a denser connective tissue matrix (hematoxylin and eosin, original magnification ×200).

    
          Figure 2.

    Figure 2. Panels illustrate two phenomena that are often assumed to be equivalent. Top, Result of a typical experimental balloon injury to a normal artery. The vessel is dilated, the media is injured, and an intima is formed. The most important result, however, is stenosis. We would define stenosis as loss of the original lumen size. Bottom, Result when a typical human atherosclerotic vessel undergoes angioplasty. In this case, the vessel is already stenotic. The balloon dilates the vessel to a lumen size assumed to be normal on the basis of a comparison with a reference vessel (usually the same artery upstream of the lesion). The angioplasty fails if the vessel simply heals, ie, returns to its preangioplasty dimension. Rather than a true stenosis, as defined above, restenosis may be thought of as a failure to remain dilated. As described in the text, we have assumed that restenosis includes a large component of intimal hyperplasia. As shown in this figure, recent cell kinetic and ultrasound studies suggest that restenosis may occur without any requirement to form new mass. Put another way, an atherosclerotic vessel that fractured during angioplasty would be considered restenotic if the fracture, like most wounds, healed. Human coronary arteries that are treated by angioplasty already have severe atherosclerotic disease and a preexisting stenosis. Clinical angioplasty produces a larger lumen; however, in ≈30% to 50% of these lesions, restenosis develops as the arterial lumen renarrows.

    
          Figure 3.

    Figure 3. Angiogram and IVUS of right coronary artery lesion treated by angioplasty. a, Tight stenosis (≈90%) of mid right coronary artery, proximal to origin of right ventricular branch (arrow). b, IVUS image of same lesion showing central placement of ultrasound catheter with minimal surrounding lumen. The echogenic inner layer and subjacent echolucent layer are generally interpreted to represent intima (large arrow) and media (small arrow), respectively. Therefore, this artery has a moderate plaque burden that is obstructing the lumen. c, Postangioplasty angiogram showing the artery to be widely patent with some residual haziness at the site of angioplasty (arrow). d, Postangioplasty IVUS image demonstrating that the echolucent arterial lumen (arrow) has been enlarged and that a moderate plaque burden remains, particularly from the 7 o’clock to 12 o’clock positions.

    Table 1. Intimal Unique Gene Expression

    Function Gene Humans Rats/Rabbits References
    In Vitro In Vivo In Vitro In Vivo
    Fetal Intimal Adult Plaque Fetal Intimal Adult Plaque Resten- osis Pup Contrac- tile Adult Syn- thetic Adult Fetal Media Intima
    Adherence Integrin α1 + +++ + +++ + ++ + +++ 182
    Adherence Integrin α3β1 + + + +++ + 182
    Adherence Integrin α5 + ++ + ++ + 182
    Adherence Integrin αVβ1 + ++ + ++++ 182
    Adherence Vitronectin ++ 258
    Adherence Metavinculin +/− + +++ + +/− +++ + 259
    Adherence Phosphoglucomutase +/− +/− + +++ + +/− +++ (V. Koteliansky, personal communication, 1994)
    Adherence 15 Lipoxygenase +++M? 260
    Angiotensin ACE + ++++1 70,261
    Angiotensin AT-1 ++++ ++ +++++ 70
    Angiotensin AT-2 + + + 70
    Coagulation PAI-1 + ++ ++ 258,262-264
    Coagulation Tissue factor ++ Transient 197,198,201
    Coagulation Thrombomodulin +++ 265
    Cytochrome P-450 CYPIA1 +++++ Inducible 189
    Cytokine Interferon Inducible Assumed present Assumed present 40
    Cytoskeleton Vinculin ++ + +++ 259,266
    Cytoskeleton SM actin +++++ +++ ++ +/− ++++ ++ +/− ++++ +++ +++++ 169,259,266-270
    Cytoskeleton Calponin + +++ + + +++ 267,271
    Cytoskeleton Cytokeratins 8 and 18 ++ +/− +++ + Focal 259,272
    Cytoskeleton Desmin + ++ ++/−− 273
    Cytoskeleton H caldesmon +/− +/− + +++ + 271,274
    Cytoskeleton Nonmuscle myosin ++ +/− ++ ++++ + +++ ++ +++ ++ 275-278
    Cytoskeleton Gelsolin 279
    Cytoskeleton SM 1 MHC ++ +++ +++ ++ + +++++ 267,280-285
    Cytoskeleton SM 2 MHC +/− + +++ ++ + +++++ 269,271,280,285
    Cytoskeleton SM 22 ++++ +++ +++++ 267
    Development HoxB7 +++++ (J.M. Miano, unpublished data, 1994)
    Development Gax ++ ++ 286
    Differentiation marker Ferritin ++++ + 287
    Growth factor bFGF +++ ++++ ++ 288,289
    Growth factor IGF-1 ++ +++ + 290-292
    Growth factor IGF-2 +++ 293
    Growth factor PDGF-A ++ +++ +++ +++ +++ + ++ 47,69,108, 109,294
    Growth factor PDGF-Rα ++++ ++++ +++ +++ 69
    Growth factor PDGF-B +++ −!! +++!! 69,294
    Immune response MHC II Inducible ++++ +++ 194,295,296
    Inflammation ICAM ++ +++ ++ ++ 44,260,297, 298
    Inflammation IL-1 Inducible ++ Inducible1 299-301
    Inflammation IL-2 + 302
    Inflammation IL-6 Inducible + + 299
    Inflammation IL-8 Inducible 303
    Inflammation MCSF-1 Inducible + +++ In rabbit and human lesions in SMCs 303-305
    Inflammation TNF-α 306,307
    Inflammation VCAM +++ ++ 44,308,309
    Inflammation c-fms Inducible ++M? 305,310
    Lipids aLDL-R Inducible +++Rabbit 310,311
    Lipids Apo(e) 312
    Lipids LPL ++M? 260,313
    Macrophage MCP-1 + 314,315
    Matrix Col I(a1)1 +++++ ?? +++++ +++++ ++ ++++? 187,192
    Matrix Elastin +++ + ++++ 267
    Matrix Elastin +++++ + + +++++ + 187,259,316, 317
    Matrix Fibronectin ED A ++++ ++++ ++++ ++++ ++++ ++++ ++++ 259
    Matrix Fibronectin ED B + + + + ++++ 259
    Matrix Gla2 +++ + +++++ 267
    Matrix Gla2 ? ++ 318
    Matrix Laminin A B1 B2 +++++ +++++ +++++ 259
    Matrix Laminin A S B2 +++++ +++++ +++++ +/− 182
    Matrix Osteopontin ++ ? ++ ++ +++ +++ ++++ ? ++ + + +++ 154,202,267
    Matrix Tenascin ++ +++++ ++ ++? ++ ++++ 184,185,319
    Matrix Versican ++ +++ +++ +++++ +++++ ? ? ? 320 (J.M. Lemire, unpublished data, 1994)
    Matrix CHIP28 +++ + +++ 267
    Membrane channel Phospholamban ++ ++ 267
    Protease Stromelysin +++ 218
    Receptor Thrombin receptor +++ +++ 0 +++ 126,127,130, 321,322
    RNA regulatory protein? F31 ? +++ +++ 293
    SR protein 1RA1 ++ 267
    TGF-β family BMP 2a +++ 203
    Unidentified sequences 2E10 +++ 267

    + indicates relative abundance; −, measurements have been made and the material in question was not present; ?, data are ambiguous; ∼, data not clear; and blanks indicate that measurements have not yet been made.

    1Contradictory data exist.

    2Different Gla proteins.

    (This table is revised and updated on a regular basis. Requests for updated reprints of the table may be addressed to Dr Schwartz.)

    Table 2. Likely Mediators of Neointimal Formation

    Description Mediators Inhibition Stimulation
    Antibody Antagonist Agonist
    First wave (0–3 days) Replication of SMCs within the media FGF + NA +
    PDGF ± NA
    TGF-β NA ND ±
    Ang II ND + ND1
    Second wave (3–14 days) Migration of SMCs from the media into the intima PDGF + NA +
    Ang II ND + ND
    FGF + NA +
    Third wave (7 days–1 mo) Proliferation of SMCs within the neointima FGF NA ±
    PDGF NA
    TGF-β NA NA +2
    Ang II ND +2

    + indicates supporting evidence; −, evidence against hypothesis; ±, weak response; NA, not available; and ND, not done.

    1Ang II does stimulate medial smooth muscle.

    2Restimulation experiments.

    This study was supported by National Institutes of Health grants HL-42270 and HL-47151 and the Pacific Foundation for Cardiovascular Research. Drs deBlois and O’Brien were Research Fellows of the Medical Research Council of Canada. The authors are indebted to Dr Victor Koteliansky for help in assembling Table 1 and extensive discussions of intimal versus medial gene expression. Other contributors to Table 1 include Drs Michael Rosenfeld, Gene Liau, and Adriana Gittenberger-de Groot. The authors are also indebted to Dr Marino Labinaz of Duke University Medical Center for his insight in imaging coronary lesions and for providing Fig 3. We appreciate the critical comments regarding clinical implications provided by Dr Douglas Stewart of the University of Washington and are grateful to Drs John Simpson and Tomoaki Hinohara of Sequoia Hospital in Redwood City, Calif, for their critical comments and collaboration in atherectomy-based research. Drs Cecilia M. Giachelli and Mark W. Majesky have been vital collaborators in the studies in this review that were performed in Dr Schwartz’s laboratory. Holly Kabinoff is deserving of special thanks for her expertise in preparing this manuscript.

    Footnotes

    Correspondence to Dr Stephen M. Schwartz, MD, PhD, Department of Pathology, University of Washington, Vascular Biology/Box 357335, Seattle, WA 98195-7335.

    References

    • 1 French JE. Atherosclerosis in relation to the structure and function of the arterial intima, with special reference to the endothelium. Int Rev Exp Pathol.1966; 5:253-354. MedlineGoogle Scholar
    • 2 Pearson TA, Solez K, Dillman J, Heptinstall R. Monoclonal characteristics of organising arterial thrombi: significance in the origin and growth of human atherosclerotic plaques. Lancet.1979; 1:7-11. CrossrefMedlineGoogle Scholar
    • 3 Pearson TA. Clonal characteristics of experimentally induced atherosclerotic lesions in the hybrid hare. Science.1979; 206:1423-1425. CrossrefMedlineGoogle Scholar
    • 4 Thomas WA, Reiner JM, Janakidevi K, Florentin RA, Lee KT. Population dynamics of arterial cells during atherogenesis, X: study of monotypism in atherosclerotic lesions of black women heterozygous for glucose-6-phosphate dehydrogenase (G-6-PD). Exp Mol Pathol.1979; 31:367-386.CrossrefMedlineGoogle Scholar
    • 5 Benditt EP, Benditt JM. Evidence for a monoclonal origin of human atherosclerotic plaques. Proc Natl Acad Sci U S A.1973; 70:1753-1756. CrossrefMedlineGoogle Scholar
    • 6 Thayer JM, Giachelli CM, Mirkes PE, Schwartz SM. Expression of osteopontin in the head process late in gastrulation in the rat. J Exp Zool.1995; 272:240-244. CrossrefMedlineGoogle Scholar
    • 7 Ruzicka D, Schwartz RJ. Sequential activation of α-actin genes during avian cardiogenesis: vascular smooth muscle α-actin gene transcripts mark the onset of cardiac myocyte differentiation. J Cell Biol.1988; 107:2575-2586. CrossrefMedlineGoogle Scholar
    • 8 Woodcock-Mitchell J, Mitchell JJ, Low RB, Kieny M, Sengel P, Rubbia L, Skalli O, Jackson B, Gabbiani G. α-Smooth muscle actin is transiently expressed in embryonic rat cardiac and skeletal muscles. Differentiation.1988; 39:161-166. CrossrefMedlineGoogle Scholar
    • 9 Schmid E, Osborn M, Rungger-Brandle E, Gabbiani G, Weber K, Franke W. Distribution of vimentin and desmin filaments in smooth muscle tissue of mammalian and avian aorta. Exp Cell Res.1982; 137:329-340. CrossrefMedlineGoogle Scholar
    • 10 Glukhova MA, Kabakov AE, Frid MG, Ornatsky OI, Belkin AM, Mukhin DN, Orekhov AN, Koteliansky VE, Smirnov VN. Modulation of human aortic smooth muscle cell phenotype: a study of muscle-specific variants of vinculin, caldesmon, and actin expression. Proc Natl Acad Sci U S A.1988; 85:9542-9546. CrossrefMedlineGoogle Scholar
    • 11 Kuro-o M, Nagai R, Tsuchimochi H, Katoh H, Yazaki Y, Ohkubo A, Takaku F. Developmentally regulated expression of vascular smooth muscle cell myosin heavy chain isoforms. J Biol Chem.1989; 264:18272-18275. CrossrefMedlineGoogle Scholar
    • 12 Frid MG, Printseva OY, Chiavegato A, Faggin E, Scatena M, Koteliansky VE, Pauletto P, Glukhova MA, Sartore S. Myosin heavy-chain isoform composition and distribution in developing and adult human aortic smooth muscle cells. J Vasc Res.1993; 30:279-292. CrossrefMedlineGoogle Scholar
    • 13 Price RJ, Owens GK, Skalak TC. Immunohistochemical identification of arteriolar development using markers of smooth muscle differentiation: evidence that capillary arterialization proceeds from terminal arterioles. Circ Res.1994; 75:520-527. CrossrefMedlineGoogle Scholar
    • 14 Velican D, Velican C. Intimal thickening in developing coronary arteries and its relevance to atherosclerotic involvement. Atherosclerosis.1976; 23:345-355. CrossrefGoogle Scholar
    • 15 Stary HC. Evolution and progression of atherosclerotic lesions in coronary arteries of children and young adults. Arteriosclerosis. 1989;9(suppl 1):119-132. Google Scholar
    • 16 Scott RF, Thomas WA, Lee WM, Reiner JM, Florentin RA. Distribution of intimal smooth muscle cells mass and their relationship to early atherosclerosis in the abdominal aortas of young swine. Atherosclerosis.1979; 34:291-301. CrossrefMedlineGoogle Scholar
    • 17 Sims FH, Gavin JB, Vanderwee MA. The intima of human coronary arteries. Am Heart J.1989; 118:32-38. CrossrefMedlineGoogle Scholar
    • 18 Slomp J, Van Munsteren JC, Poelmann RE, DeReeder EG, Bogers AJJC, Gittenberger-de Groot AC. Formation of intimal cushions in the ductus arteriosus as a model for vascular intimal thickening: an immunohistochemical study of changes in extracellular matrix components. Atherosclerosis.1992; 93:25-39. CrossrefMedlineGoogle Scholar
    • 19 Boudreau N, Turley E, Rabinovitch M. Fibronectin, hyaluronan, and a hyaluronan binding protein contribute to increased ductus arteriosus smooth muscle cell migration. Dev Biol.1991; 143:235-247. CrossrefMedlineGoogle Scholar
    • 20 Chobanian AV, Prescott MF, Haudenschild CC. Recent advances in molecular pathology: the effects of hypertension on the arterial wall. Exp Mol Pathol.1984; 41:153-169. CrossrefMedlineGoogle Scholar
    • 21 Friedman RJ, Stemerman MB, Wenz B, Moore S, Gauldie J, Gent M, Tiell ML, Spaet TH. The effect of thrombocytopenia on experimental atherosclerotic lesion formation in rabbits: smooth muscle cell proliferation and re-endothelialization. J Clin Invest.1977; 60:1191-1201. CrossrefMedlineGoogle Scholar
    • 22 Bjorkerud S, Bondjers G. Arterial repair and atherosclerosis after mechanical injury: tissue response after induction of a large superficial transverse injury. Atherosclerosis. 1973;18(pt 5):235-255. Google Scholar
    • 23 Schwartz SM, Stemerman MB, Benditt EP. The aortic intima, II: repair of the aortic lining after mechanical denudation. Am J Pathol.1975; 81:15-42. MedlineGoogle Scholar
    • 24 Clowes AW, Reidy MA, Clowes MM. Mechanisms of stenosis after arterial injury. Lab Invest.1983; 49:208-215. MedlineGoogle Scholar
    • 25 Betz E, Schlote W. Response of vessel walls to chronically applied electrical stimuli. Basic Res Cardiol.1979; 74:10-20. CrossrefMedlineGoogle Scholar
    • 26 Baumgartner HR. Eine neue Methode zur Erzeugung der Thromben durch gezielte Uberdehnung der Gefasswand. Z Ges Exp Med.1963; 137:227-234. CrossrefMedlineGoogle Scholar
    • 27 Stemerman MB, Spaet TH, Pitlick F, Clintron J, Lejneks I, Tiell ML. Intimal healing: the pattern of reendothelialization and intimal thickening. Am J Pathol.1977; 87:125-142. MedlineGoogle Scholar
    • 28 Nomoto A, Hirosumi J, Sekiguchi C, Mutoh S, Yamaguchi I, Aoki H. Antiatherogenic activity of FR34235 (Nilvadapine), a new potent calcium antagonist: effect on cuff-induced intimal thickening of rabbit carotid artery. Atherosclerosis.1987; 64:255-261. CrossrefMedlineGoogle Scholar
    • 29 Haudenschild C, Studer A. Early interactions between blood cells and severely damaged rabbit aorta. Eur J Clin Invest.1971; 2:1-7. CrossrefMedlineGoogle Scholar
    • 30 Baumgartner HR. Effects of acetylsalicylic acid, sulfinpyrazone and dipridamole on platelet adhesion and aggregation in flowing native and anti-coagulated blood. Haemostasis.1979; 8:340-352. MedlineGoogle Scholar
    • 31 Schwartz SM, Reidy MA, Clowes A. Kinetics of atherosclerosis: a stem cell model. Ann N Y Acad Sci.1985; 454:292-304. CrossrefMedlineGoogle Scholar
    • 32 Clowes AW, Clowes MM, Reidy MA. Kinetics of cellular proliferation after arterial injury, I: smooth muscle growth in the absence of endothelium. Lab Invest.1983; 49:327-333. MedlineGoogle Scholar
    • 33 Clowes AW, Schwartz SM. Significance of quiescent smooth muscle migration in the injured rat carotid artery. Circ Res.1985; 56:139-145. CrossrefMedlineGoogle Scholar
    • 34 Hanke H, Strohschneider T, Oberhoff M, Betz E, Karsch KR. Time course of smooth muscle cell proliferation in the intima and media of arteries following experimental angioplasty. Circ Res.1990; 67:651-659. CrossrefMedlineGoogle Scholar
    • 35 Vesselinovitch D. Animal models and the study of atherosclerosis. Arch Pathol Lab Med.1988; 112:1011-1017. MedlineGoogle Scholar
    • 36 Anderson PG. Restenosis: animal models and morphometric techniques in studies of the vascular response to injury. Cardiovasc Pathol.1992; 1:263-278.CrossrefMedlineGoogle Scholar
    • 37 Salisbury BG, Hajjar DP, Minick CR. Altered glycosaminoglycan metabolism in injured arterial wall. Exp Mol Pathol.1985; 42:306-319. CrossrefMedlineGoogle Scholar
    • 38 Schwartz SM, Haudenschild CC, Eddy EM. Endothelial regeneration, I: quantitative analysis of initial stages of endothelial regeneration in rat aortic intima. Lab Invest.1978; 38:568-580. MedlineGoogle Scholar
    • 39 Reidy MA, Yoshida K, Harker LA, Schwartz SM. Vascular injury: quantification of experimental focal endothelial denudation in rats using indium-111–labeled platelets. Arteriosclerosis.1986; 6:305-311. LinkGoogle Scholar
    • 40 Jonasson L, Holm J, Hansson GK. Smooth muscle cells express Ia antigens during arterial response to injury. Lab Invest.1988; 58:310-315. MedlineGoogle Scholar
    • 41 Bjorkerud S. Reaction of the aortic wall of the rabbit after superficial, longitudinal, mechanical trauma. Virchows Arch.1969; 347:197-210. CrossrefGoogle Scholar
    • 42 Schwartz RS, Edwards WD, Huber KC, Antoniades LC, Bailey KR, Camrud AR, Jorgenson MA, Holmes DR. Coronary restenosis: prospects for solution and new perspectives from a porcine model. Mayo Clin Proc.1993; 68:54-62. CrossrefMedlineGoogle Scholar
    • 43 Weidinger FF, McLenachan JM, Cybulsky MI, Fallon JT, Hollenberg NK, Cooke JP, Ganz P. Hypercholesterolemia enhances macrophage recruitment and dysfunction of regenerated endothelium after balloon injury of the rabbit iliac artery. Circulation.1991; 84:755-767. CrossrefMedlineGoogle Scholar
    • 44 Tanaka H, Sukhova GK, Swanson SJ, Clinton SK, Ganz P, Cybulsky MI, Libby P. Sustained activation of vascular cells and leukocytes in the rabbit aorta after balloon injury. Circulation.1993; 88:1788-1803. CrossrefMedlineGoogle Scholar
    • 45 Lindner V, Reidy MA. Proliferation of smooth muscle cells after vascular injury is inhibited by an antibody against basic fibroblast growth factor. Proc Natl Acad Sci U S A.1991; 88:3739-3743. CrossrefMedlineGoogle Scholar
    • 46 Olson NE, Chao S, Lindner V, Reidy MA. Intimal smooth muscle cell proliferation after balloon catheter injury: the role of basic fibroblast growth factor. Am J Pathol.1992; 140:1017-1023. MedlineGoogle Scholar
    • 47 Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature.1993; 362:801-813. CrossrefMedlineGoogle Scholar
    • 48 Jawien A, Bowen-Pope DF, Lindner V, Schwartz SM, Clowes AW. Platelet-derived growth factor promotes smooth muscle migration and intimal thickening in a rat model of balloon angioplasty. J Clin Invest.1992; 89:507-511. CrossrefMedlineGoogle Scholar
    • 49 Ferns GAA, Raines EW, Sprugel KH, Motani AS, Reidy MA, Ross R. Inhibition of neointimal smooth muscle accumulation after angioplasty by an antibody to PDGF. Science.1991; 253:1129-1132. CrossrefMedlineGoogle Scholar
    • 50 Jackson CL, Raines EW, Ross R, Reidy MA. Role of endogenous platelet-derived growth factor in arterial smooth muscle cell migration after balloon catheter injury. Arterioscler Thromb.1993; 13:1218-1226. CrossrefMedlineGoogle Scholar
    • 51 Daemen MJAP, Lombardi DM, Bosman FT, Schwartz SM. Angiotensin II induces smooth muscle cell proliferation in the normal and injured rat arterial wall. Circ Res.1991; 68:450-456.CrossrefMedlineGoogle Scholar
    • 52 van Kleef EM, Smits JFM, De Mey JGR, Cleutjens JPM, Lombardi DM, Schwartz SM, Daemen MJAP. α1-Adrenoreceptor blockade reduces the angiotensin II–induced vascular smooth muscle cell DNA synthesis in the rat thoracic aorta and carotid artery. Circ Res.1992; 70:1122-1127. CrossrefMedlineGoogle Scholar
    • 53 Prescott M, Webb R, Reidy MA. ACE inhibitors vs AII, AT1 receptor antagonist: effects on smooth muscle cell migration and proliferation after balloon catheter injury. Am J Pathol.1991; 139:1291-1302. MedlineGoogle Scholar
    • 54 Bennett MR, Schwartz SM. Antisense therapy for angioplasty restenosis: some critical considerations. Circulation. In press. Google Scholar
    • 55 Ebbecke M, Unterberg C, Buchwald A, Stohr S, Wiegand V. Antiproliferative effects of a c-myc antisense oligonucleotide on human arterial smooth muscle cells. Basic Res Cardiol.1992; 87:585-591. CrossrefMedlineGoogle Scholar
    • 56 Shi Y, Fard A, Galeo A, Hutchinson HG, Vermani P, Dodge GR, Hall DJ, Shaheen F, Zalewski A. Transcatheter delivery of c-myc antisense oligomers reduces neointimal formation in a porcine model of coronary artery balloon injury. Circulation.1994; 90:944-951. CrossrefMedlineGoogle Scholar
    • 57 Simons M, Edelman ER, Rosenberg RD. Antisense proliferating cell nuclear antigen oligonucleotides inhibit intimal hyperplasia in a rat carotid artery injury model. J Clin Invest.1994; 93:2351-2356. CrossrefMedlineGoogle Scholar
    • 58 Morishita R, Gibbons GH, Ellison KE, Nakajima M, von der Leyen H, Zhang L, Kaneda Y, Ogihara T, Dzau VJ. Intimal hyperplasia after vascular injury is inhibited by antisense cdk 2 kinase oligonucleotides. J Clin Invest.1994; 93:1458-1464. CrossrefMedlineGoogle Scholar
    • 59 Abe J, Zhou W, Taguchi J, Takuwa N, Miki K, Okazaki H, Kurokawa K, Kumada M, Takuwa Y. Suppression of neointimal smooth muscle cell accumulation in vivo by antisense cdc2 and cdk2 oligonucleotides in rat carotid artery. Biochem Biophys Res Commun.1994; 198:16-24. CrossrefMedlineGoogle Scholar
    • 60 Biro S, Fu YM, Yu ZX, Epstein SE. Inhibitory effects of antisense oligodeoxynucleotides targeting c-myc mRNA on smooth muscle cell proliferation and migration. Proc Natl Acad Sci U S A.1993; 90:654-658. CrossrefMedlineGoogle Scholar
    • 61 Majesky MW, Lindner V, Twardzik DR, Schwartz SM, Reidy MA. Production of transforming growth factor β-1 during repair of arterial injury. J Clin Invest.1991; 88:904-910. CrossrefMedlineGoogle Scholar
    • 62 Fingerle J, Johnson R, Clowes AW, Majesky MW, Reidy MA. Role of platelets in smooth muscle cell proliferation and migration after vascular injury in rat carotid artery. Proc Natl Acad Sci U S A.1989; 86:8412-8416. CrossrefMedlineGoogle Scholar
    • 63 Floege J, Eng E, Young BA, Alpers CE, Barrett TB, Bowen-Pope DF, Johnson RJ. Infusion of platelet-derived growth factor or basic fibroblast growth factor induces selective glomerular mesangial cell proliferation matrix accumulation in rats. J Clin Invest.1993; 92:2952-2962. CrossrefMedlineGoogle Scholar
    • 64 Clowes AW, Clowes MM, Reidy MA. Kinetics of cellular proliferation after arterial injury, III: endothelial and smooth muscle growth in chronically denuded vessels. Lab Invest.1986; 54:295-303. MedlineGoogle Scholar
    • 65 Nabel EG, Yang Z, Liptay S, San H, Gordon D, Haudenschild CC, Nabel GJ. Recombinant platelet-derived growth factor B gene expression in porcine arteries induce intimal hyperplasia in vivo. J Clin Invest.1993; 91:1822-1829. CrossrefMedlineGoogle Scholar
    • 66 Vassbotn FS, Ostman A, Langeland N, Holmsen H, Westermark N, Heldin CH, Nister M. Activated platelet-derived growth factor autocrine pathway transformed phenotype of a human glioblastoma cell line. J Cell Physiol.1994; 158:381-389. CrossrefMedlineGoogle Scholar
    • 67 LaRochelle WJ, Giese N, May-Siroff M, Robbins KC, Aaronson SA. Molecular localization of the transforming and secretory proteins of PDGF-A and PDGF-B. Science.1990; 248:1541-1544. CrossrefMedlineGoogle Scholar
    • 68 Pech M, Gazit A, Arnstein P, Aaronson SA. Generation of fibrosarcomas in vivo by a retrovirus that expresses the normal B chain of platelet-derived growth factor and mimicks the alternative splice pattern of the v-sis oncogene. Proc Natl Acad Sci U S A.1989; 86:2693-2697. CrossrefMedlineGoogle Scholar
    • 69 Majesky MW, Reidy MA, Bowen-Pope DF, Wilcox JN, Schwartz SM. Platelet-derived growth factor (PDGF) ligand and receptor gene expression during repair of arterial injury. J Cell Biol.1990; 111:2149-2158. CrossrefMedlineGoogle Scholar
    • 70 Viswanathan M, Stromberg C, Seltzer A, Saavedra JM. Balloon angioplasty enhances the expression of angiotensin II AT1 receptors in neointima of rat aorta. J Clin Invest.1992; 90:1707-1712. CrossrefMedlineGoogle Scholar
    • 71 Cercek B, Fishbein MC, Forrester JS, Helfant RH, Fagin JA. Induction of insulin-like growth factor I messenger RNA in rat aorta after balloon denudation. Circ Res.1990; 66:1755-1760. LinkGoogle Scholar
    • 72 Haudenschild CC, Schwartz SM. Endothelial regeneration, II: restitution of endothelial continuity. Lab Invest.1979; 41:407-418. MedlineGoogle Scholar
    • 73 Garg UC, Hassid A. Nitric oxide-generating vasodilators and 8-bromo-cyclic guanosine monophosphate inhibit mitogenesis and proliferation of cultured rat vascular smooth muscle cells. J Clin Invest.1989; 83:1774-1777. CrossrefMedlineGoogle Scholar
    • 74 McNamara DB, Bedi B, Aurora H, Tena L, Ignarro LJ, Kadowitz PJ, Akers DL. L-Arginine inhibits balloon catheter-induced intimal hyperplasia. Biochem Biophys Res Commun.1993; 193:291-296. CrossrefMedlineGoogle Scholar
    • 75 Hamon M, Vallet B, Bauters C, Wernert N, McFadden EP, Lablance JM, Dupuis B, Bertrand ME. Long-term oral administration of l-arginine reduces intimal thickening and enhances neoendothelium-dependent acetylcholine-induced relaxation after arterial injury. Circulation.1994; 90:1357-1362. CrossrefMedlineGoogle Scholar
    • 76 Tarry WC, Makhoul RG. L-Arginine improves endothelium-dependent vasorelaxation and reduces intimal hyperplasia after balloon angioplasty. Arterioscler Thromb.1994; 14:938-943. CrossrefMedlineGoogle Scholar
    • 77 Farhy RD, Carretero OA, Ho K-L, Scicli AG. Role of kinins and nitric oxide in the effects of angiotensin converting enzyme inhibitors on neointima formation. Circ Res.1993; 72:1202-1210. CrossrefMedlineGoogle Scholar
    • 78 Hansson GK, Geng YJ, Holm J, Hardhammar P, Wennmalm A, Jennische E. Arterial smooth muscle cells express nitric oxide synthase in response to endothelial injury. J Exp Med.1994; 180:733-738. CrossrefMedlineGoogle Scholar
    • 79 Hassid A, Arabshahi H, Bourcier T, Dhaunsi GS, Matthews C. Nitric oxide selectively amplifies FGF-2-induced mitogenesis in primary rat aortic smooth muscle cells. Am J Physiol.1994; 267:H1040-H1048. MedlineGoogle Scholar
    • 80 Powell J, Clozel J, Muller R, Kuhn H, Hefti F, Hosang M, Baumgartner H. Inhibitors of angiotensin-converting enzyme prevent myointimal proliferation after vascular injury. Science.1989; 245:186-188. CrossrefMedlineGoogle Scholar
    • 81 MERCATOR study group. Does the new angiotensin-converting enzyme inhibitor cilazapril prevent restenosis after percutaneous transluminal coronary angioplasty? Circulation.1992; 86:100-110. CrossrefMedlineGoogle Scholar
    • 82 Kaartinen M, Penttilä A, Kovanen PT. Mast cells of two types differing in neutral protease composition in the human aortic intima: demonstration of tryptase- and tryptase/chymase-containing mast cells in normal intimas, fatty streaks, and the shoulder region of atheromas. Arterioscler Thromb.1994; 14:966-972. CrossrefMedlineGoogle Scholar
    • 83 Pan X, Nelken N, Colyvas N, Rapp JH. Inhibition of injury-induced intimal hyperplasia by saralasin in rats. J Vasc Surg.1992; 15:693-698. CrossrefMedlineGoogle Scholar
    • 84 Laporte S, Escher E. Neointima formation after vascular injury is angiotensin II-mediated. Biochem Biophys Res Commun.1991; 187:1510-1516. Google Scholar
    • 85 Dzau VJ, Gibbons GH, Pratt RE. Molecular mechanisms of vascular renin-angiotensin system in myointimal hyperplasia. Hypertension. 1991;18(suppl II):II-100-II-105. Google Scholar
    • 86 Campbell DJ. Extrarenal renin and blood pressure regulation: an alternative viewpoint. Hypertension.1989; 2:266-275. CrossrefGoogle Scholar
    • 87 Viswanathan M, Seltzer A, Saavedra JM. Heterogeneous expression of angiotensin II AT1 receptors in neointima of rat carotid artery and aorta after balloon catheter injury. Peptides.1994; 15:1205-1212. CrossrefMedlineGoogle Scholar
    • 88 Rakugi H, Jacob HJ, Krieger JE, Ingelfinger JR, Pratt RE. Vascular injury induces angiotensinogen gene expression in the media and neointima. Circulation.1993; 87:283-290. CrossrefMedlineGoogle Scholar
    • 89 Campbell DJ, Habener JF. Angiotensinogen gene is expressed and differentially regulates multiple tissues of the rat. J Clin Invest.1986; 78:31-39. CrossrefMedlineGoogle Scholar
    • 90 Cassis LA, Lynch KR, Peach MJ. Localization of angiotensinogen messenger RNA in rat aorta. Circ Res.1988; 62:1259-1262. CrossrefMedlineGoogle Scholar
    • 91 Rosenthal J, Thurnreiter M, Plaschke M, Geyer M, Reiter W, Dahlheim H. Reninlike enzymes in human vasculature. Hypertension.1990; 15:848-853. LinkGoogle Scholar
    • 92 Rogerson FM, Chai SY, Schlawe I, Murray WK, Marley PD, Mendelsohn FA. Presence of angiotensin converting enzyme in the adventitia of large blood vessels. J Hypertens.1992; 10:615-620. CrossrefMedlineGoogle Scholar
    • 93 Fishel RS, Thourani V, Shai SY, Corson MA, Nabel EG, Bernstein KE, Berk BC. Fibroblast growth factor stimulates angiotensin converting enzyme expression in vascular smooth muscle cells: possible mediator of the response to vascular injury. J Clin Invest..1995; 95:377-387. CrossrefMedlineGoogle Scholar
    • 94 Shiota N, Okunishi H, Fukamizu A, Sakonjo H, Kikumori M, Nishimura T, Nakagawa T, Murakami K, Miyazaki M. Activation of two angiotensin-generating systems in the balloon-injured artery. FEBS Lett.1993; 323:239-242. CrossrefMedlineGoogle Scholar
    • 95 Janiak P, Pillon A, Prost J, Vilaine J. Role of angiotensin subtype 2 receptor in neointima formation after vascular injury. Hypertension.1992; 20:737-745. LinkGoogle Scholar
    • 96 Ohishi M, Fujii K, Minamino T, Higaki J, Kamitani A, Zhao Y, Mikami H, Miki T, Ogihava T. A potent genetic risk factor for restenosis. Nat Genet.1993; 5:324-325. CrossrefMedlineGoogle Scholar
    • 97 Cambien F, Poirier O, Lecerf L, Evans A, Cambou JP, Arveiler D, Luc G, Bard JM, Bara L, Ricard S. Deletion polymorphism in the gene for angiotensin-converting enzyme is a potent risk factor for myocardial infarction. Nature.1992; 359:641-644. CrossrefMedlineGoogle Scholar
    • 98 Lam JYT, Lacoste L, Bourassa MG. Cilazapril and early atherosclerotic changes after balloon injury of porcine carotid arteries. Circulation.1992; 85:1542-1547. CrossrefMedlineGoogle Scholar
    • 99 Huber KC, Schwartz RS, Edwards WD, Camrud AR, Bailey KR, Jorgenson MA, Holmes DR. Effects of angiotensin converting enzyme inhibition on neointimal proliferation in a porcine coronary injury model. Am Heart J.1993; 125:695-701. CrossrefMedlineGoogle Scholar
    • 100 Hanson SR, Powell JS, Dodson T, Lumsden A, Kelly AB, Anderson JS, Clowes AW, Harker LA. Effects of angiotensin-converting enzyme inhibition with cilazapril on intimal hyperplasia in injured arteries and vascular grafts in the baboon. Hypertension. 1991;18(suppl II):II-70-II-76. Google Scholar
    • 101 Urata H, Ganten D. Cardiac angiotensin II formation: the angiotensin I converting enzyme and human chymase. Eur Heart J. 1993;14(suppl I):177-182. Google Scholar
    • 102 Reilly CF, Tewksbury DA, Schechter NM, Travis J. Rapid conversion of angiotensin I to angiotensin II by neutrophil and mast cell proteinases. J Biol Chem.1982; 257:8619-8622. CrossrefMedlineGoogle Scholar
    • 103 Snyder RA, Watt KW, Wintroub BU. A human platelet angiotensin I-processing system: identification of components and inhibition of angiotensin-converting enzyme byproduct. J Biol Chem.1985; 260:7857-7860. CrossrefMedlineGoogle Scholar
    • 104 Johnson H, Drummer OH. Hydrolysis of angiotensin I by peptidases in homogenates of rat lung and aorta. Biochem Pharmacol.1988; 37:1131-1136. CrossrefMedlineGoogle Scholar
    • 105 Walker L, Bowen-Pope DF, Ross R, Reidy MA. Production of PDGF-like molecules by cultured arterial smooth muscle cells accompanies proliferation after arterial injury. Proc Natl Acad Sci U S A.1986; 83:7311-7315. CrossrefMedlineGoogle Scholar
    • 106 Seifert RA, Schwartz SM, Bowen-Pope DF. Developmentally regulated production of platelet-derived growth factor-like molecules. Nature.1984; 311:669-671. CrossrefMedlineGoogle Scholar
    • 107 Lindner V, Giachelli CM, Schwartz SM, Reidy MA. A subpopulation of smooth muscle cells in rat arteries express PDGF–B chain mRNA. Circ Res.1995; 76:951-957. CrossrefMedlineGoogle Scholar
    • 108 Libby P, Warner SJ, Salomon RN, Birinyi LK. Production of platelet-derived growth factor-like mitogen by smooth-muscle cells from human atheroma. N Engl J Med.1988; 318:1493-1498. CrossrefMedlineGoogle Scholar
    • 109 Wilcox JN, Smith KM, Williams LT, Schwartz SM, Gordon D. Platelet-derived growth factor mRNA detection in human atherosclerotic plaques by in situ hybridization. J Clin Invest.1988; 82:1134-1143. CrossrefMedlineGoogle Scholar
    • 110 Rekhter MD, Gordon D. Does platelet-derived growth factor-A chain stimulate proliferation of arterial mesenchymal cells in human atherosclerotic plaques? Circ Res.1994; 75:410-417. CrossrefMedlineGoogle Scholar
    • 111 Nabel EG, Shum L, Pompili VJ, Yang ZY, San H, Shu HB, Liptay S, Gold L, Gordon D, Derynck R. Direct transfer of transforming growth factor-β1 gene into arteries stimulates fibrocellular hyperplasia. Proc Natl Acad Sci U S A.1993; 90:10759-10763. CrossrefMedlineGoogle Scholar
    • 112 Wolf YG, Rasmussen LM, Ruoslahti E. Antibodies against transforming growth factor-β1 suppress intimal hyperplasia in a rat model. J Clin Invest.1994; 93:1172-1178. CrossrefMedlineGoogle Scholar
    • 113 Nikol S, Isner JM, Pickering JG, Kearney M, Leclerc G, Weir L. Expression of transforming growth factor-β1 is increased in human vascular restenosis lesions. J Clin Invest.1992; 90:1582-1592. CrossrefMedlineGoogle Scholar
    • 114 Grainger DJ, Kirschenlohr HL, Metcalfe JC, Weissberg PL. Proliferation of human smooth muscle cells promoted by lipoprotein(a). Science.1993; 260:1655-1658. CrossrefMedlineGoogle Scholar
    • 115 Majack RA, Majesky MW, Goodman LV. Role of PDGF-A expression in the control of vascular smooth muscle cell growth by transforming growth factor-β. J Cell Biol.1990; 111:239-247. CrossrefMedlineGoogle Scholar
    • 116 Battegay E, Raines E, Seifert RA, Bowen-Pope DF, Ross R. TGF-β induces bimodal proliferation of connective tissue cells via complex control of an autocrine PDGF loop. Cell.1990; 63:515-524. CrossrefMedlineGoogle Scholar
    • 117 Gibbons G, Pratt R, Dzau V. Vascular smooth muscle cell hypertrophy vs hyperplasia: autocrine TGF-β1 expression determines growth response to angiotensin II. J Clin Invest.1992; 90:456-461. CrossrefMedlineGoogle Scholar
    • 118 Tenda K, Saikawa T, Maeda T, Sato Y, Niwa H, Inoue T, Yonemochi H, Maruyama T, Shimoyama N, Aragaki S. The relationship between serum lipoprotein(a) and restenosis after initial elective percutaneous transluminal coronary angioplasty. Jpn Circ J.1993; 57:789-795. CrossrefMedlineGoogle Scholar
    • 119 Hearn JA, Donohue BC, Ba’albaki H, Douglas JS, King SBI, Lembo NJ, Roubin GS, Sgoutas DS. Usefulness of serum lipoprotein(a) as a predictor of restenosis after percutaneous transluminal coronary angioplasty. Am J Cardiol.1992; 69:736-739. CrossrefMedlineGoogle Scholar
    • 120 Ezratty A, Simon DI, Loscalzo J. Lipoprotein(a) binds to human platelets and attenuates plasminogen binding activation. Biochemistry.1993; 32:4628-4633. CrossrefMedlineGoogle Scholar
    • 121 Etingin OR, Hajjar DP, Hajjar KA, Harpel PC, Nachman RL. Lipoprotein(a) regulates plasminogen activator inhibitor-1 expression in endothelial cells: a potential mechanism in thrombogenesis. J Biol Chem.1991; 266:2459-2465. CrossrefMedlineGoogle Scholar
    • 122 Sporn MB, Roberts AB, Wakefield LM, De Crombrugghe B. Some recent advances in the chemistry and biology of transforming growth factor-β. J Cell Biol.1987; 105:1039-1046. CrossrefMedlineGoogle Scholar
    • 123 Kirschenlohr HL, Metcalfe JC, Weissberg PL, Grainger DJ. Adult human aortic smooth muscle cells in culture produce active TGF-β. Am J Physiol.1993; 265:C571-C576. CrossrefMedlineGoogle Scholar
    • 124 Grainger DJ, Weissberg PL, Metcalfe JC. Tamoxifen decreases the rate of proliferation of rat vascular smooth muscle cells in culture by inducing production of transforming growth factor-β. Biochem J.1993; 294:109-112. CrossrefMedlineGoogle Scholar
    • 125 Grainger DJ, Kemp PR, Liu AC, Lawn RM, Metcalfe JC. Activation of transforming growth factor-β is inhibited in transgenic apolipoprotein(a) mice. Nature.1994; 370:460-462. CrossrefMedlineGoogle Scholar
    • 126 Schwartz SM. Serum-derived growth factor is thrombin. J Clin Invest.1993; 91:4. CrossrefMedlineGoogle Scholar
    • 127 McNamara CA, Sarembock IJ, Gimple LW, Fenton JWI, Coughlin SR, Owens GK. Thrombin stimulates proliferation of cultured rat aortic smooth muscle cells by a proteolytically activated receptor. J Clin Invest.1993; 91:94-98. CrossrefMedlineGoogle Scholar
    • 128 Hatton MW, Moar SL, Richardson M. De-endothelialization in vivo initiates a thrombogenic reaction at the rabbit aorta surface. Am J Pathol.1989; 135:499-508. MedlineGoogle Scholar
    • 129 Pasche B, Swedenborg J, Hedin U, Olsson P, Ljungqvist A. Thrombin-inhibitory capacity of the injured vessel wall. Thromb Res.1991; 62:531-544. CrossrefMedlineGoogle Scholar
    • 130 Nelken NA, Soifer SJ, O’Keefe J, Vu TK, Charo IF, Coughlin SR. Thrombin receptor expression in normal and atherosclerotic human arteries. J Clin Invest.1992; 90:1614-1621. CrossrefMedlineGoogle Scholar
    • 131 Hong MK, Bhatti T, Matthews BJ, Stark KS, Cathapermal SS, Foegh ML, Ramwell PW, Kent KM. The effect of porous infusion balloon-delivered angiopeptin on myointimal hyperplasia after balloon injury in the rabbit. Circulation.1993; 88:638-648. CrossrefMedlineGoogle Scholar
    • 132 Santoian ED, Schneider JE, Gravanis MB, Foegh M, Tarazona N, Cipolla GD, King SBI. Angiopeptin inhibits intimal hyperplasia after angioplasty in porcine coronary arteries. Circulation.1993; 88:11-14. CrossrefMedlineGoogle Scholar
    • 133 Lundergan C, Foegh ML, Vargas R, Eufemio M, Bormes GW, Kot PA, Ramwell PW. Inhibition of myointimal proliferation of the rat carotid artery by the peptides, angiopeptin and BIM 23034. Atherosclerosis.1989; 80:49-55. CrossrefMedlineGoogle Scholar
    • 134 Foegh ML, Khirabadi BS, Chambers E, Amamoo S, Ramwell PW. Inhibition of coronary artery transplant atherosclerosis in rabbits with angiopeptin, an octapeptide. Atherosclerosis.1989; 78:229-236. CrossrefMedlineGoogle Scholar
    • 135 Calcagno D, Conte JV, Howell MH, Foegh ML. Peptide inhibition of neointimal hyperplasia in vein grafts. J Vasc Surg.1991; 13:475-479. CrossrefMedlineGoogle Scholar
    • 136 Emanuelsson H, Bagger J, Balcon R, Beatt K, Foegh M, Piessens J, Popma JJ. Long-term effects of angiopeptin treatment in coronary angioplasty: reduction of clinical events but not of angiographic restenosis. J Am Coll Cardiol. 1994;59A. Abstract. Google Scholar
    • 137 Neff M, M’etry JM, Frick P, Anliker M, Knoblauch M. The microvasculature of the small intestinal mucosa of the rat: quantification of hemodynamic effects of topically applied cimetidine, ranitidine, somatostatin, and vasopressin. Scand J Gastroenterol. 1985;112(suppl):6-11. Google Scholar
    • 138 Long JB, Rigamonti DD, Dosaka K, Kraimer JM, Martinez-Arizala A. Somatostatin causes vasoconstriction, reduces blood flow and increases vascular permeability in the rat central nervous system. J Pharmacol Exp Ther.1992; 260:1425-1432. MedlineGoogle Scholar
    • 139 Sieber CC, Mosca PG, Groszmann RJ. Effect of somatostatin on mesenteric vascular resistance in normal and portal hypertensive rats. Am J Physiol.1992; 262:G274-G277. MedlineGoogle Scholar
    • 140 Light JT, Bellan JA, Chen IL, Longenecker LL, Murphy WA, Coy DH, Kadowitz PJ, McNamara DB. Angiopeptin enhances acetylcholine-induced relaxation and inhibits intimal hyperplasia after vascular injury. Am J Physiol.1993; 265:H1265-H1274. MedlineGoogle Scholar
    • 141 Au YP, Kenagy RD, Clowes MM, Clowes AW. Mechanisms of inhibition by heparin of vascular smooth muscle cell proliferation and migration. Haemostasis. 1993;23(suppl 1):177-182. Google Scholar
    • 142 Simons M, Edelman ER, DeKeyser J, Langer R, Rosenberg RD. Antisense c-myb oligonucleotides inhibit intimal arterial smooth muscle cell accumulation in vivo. Nature.1992; 359:67-70. CrossrefMedlineGoogle Scholar
    • 143 Ellis SG, Roubin GS, Wilentz J, Douglas JS, King SB. Effect of 18- and 24-hour heparin administration for prevention of restenosis after uncomplicated coronary angioplasty. Am Heart J.1989; 117:777-782. CrossrefMedlineGoogle Scholar
    • 144 Lehmann KG, Doria RJ, Feuer JM, Hall PX, Hoang DT. Paradoxical increase in restenosis rate with chronic heparin use: final results of a randomized trial. J Am Coll Cardiol.1991; 17:181A. Abstract. CrossrefGoogle Scholar
    • 145 Gold HK, Torres FW, Garabedian HD, Werner W, Jang I, Khan A, Hagstrom JN, Asuda TY, Leinbach RC, Newell JB, Bovill EG, Stump DC, Collen D. Evidence for a rebound coagulation phenomenon after cessation of a four-hour infusion of a specific thrombin inhibitor in patients with unstable angina pectoris. J Am Coll Cardiol.1993; 21:1039-1047. CrossrefMedlineGoogle Scholar
    • 146 Edelman ER, Karnovsky MJ. Contrasting effects of the intermittent and continuous administration of heparin in experimental restenosis. Circulation.1994; 89:770-776. CrossrefMedlineGoogle Scholar
    • 147 Faxon D, Sprio T, Minor S, Douglas J, Cote G, Dorosti K, Gottlieb R, Califf R, Topol E, Gordon J. Enoxaparin, a low molecular weight heparin, in the prevention of restenosis after angioplasty: results of a double-blind randomized trial. J Am Coll Cardiol.1992; 19:258A. Abstract. CrossrefMedlineGoogle Scholar
    • 148 Tcheng JE, Topol EJ, Kleiman NS, Ellis SG, Navetta FI, Fintel DJ, Weisman HF, Anderson K, Wang AL, Miller JA, Sigmon KN, Califf RM, EPIC investigators. Improvement in clinical outcomes of coronary angioplasty by treatment with the GPIIb/IIIa inhibitor chimeric 7E3: multivariate analysis of the EPIC study. Circulation. 1993;88(suppl I, pt 2):I-506. Abstract. Google Scholar
    • 149 Tcheng JE, Ellis S, Kleiman NS, Harrington RA, Mick MJ, Navetta FI, Worley S, Smith JE, Kereiakes DJ, Kitt MM, Miller JA, Sigmon KN, Califf RM, Topol EJ. Outcome of patients treated with the GPIIB/IIIa inhibitor Integrelin during coronary angioplasty: results of the IMPACT study. Circulation. 1993;88(suppl I, pt 2):I-595. Abstract. Google Scholar
    • 150 EPIC investigators. Use of a monoclonal antibody directed against the platelet glycoprotein IIB/IIIa receptor in high-risk coronary angioplasty. N Engl J Med.1994; 330:956-1007. CrossrefMedlineGoogle Scholar
    • 151 Choi ET, Engel L, Callow AD, Sun S, Trachtenberg J, Santoro S, Ryan US. Inhibition of neointimal hyperplasia by blocking αvβ3 integrin with a small peptide antagonist GpenGRGDSPCA. J Vasc Surg.1994; 19:125-134. CrossrefMedlineGoogle Scholar
    • 152 Liaw L, Skinner MP, Raines EW, Ross R, Cheresh DA, Schwartz SM, Giachelli CM. The adhesive and migratory effects of osteopontin are mediated via distinct cell surface integrins: role of αvβ3 in smooth muscle migration to osteopontin in vitro. J Clin Invest.1995; 95:713-724. CrossrefMedlineGoogle Scholar
    • 153 Hirota S, Imakita M, Kohri K, Ito A, Morii E, Adachi S, Kim H, Kitamura Y, Yutani C, Nomura S. Expression of osteopontin messenger RNA by macrophages in atherosclerotic plaques: a possible association with calcification. Am J Pathol.1993; 143:1003-1008. MedlineGoogle Scholar
    • 154 O’Brien ER, Garvin MR, Stewart DK, Hinohara T, Simpson JB, Schwartz SM, Giachelli CM. Osteopontin is synthesized by macrophage, smooth muscle and endothelial cells in primary and restenotic human coronary atherosclerotic plaques. Arterioscler Thromb.1994; 14:1648-1656. CrossrefMedlineGoogle Scholar
    • 155 Stary HC, Blankenhorn DH, Chandler AB, Glagov S, Insull WJ Jr, Richardson M, Rosenfeld ME, Schaffer SA, Schwartz CJ, Wagner WD, Wissler RW. A definition of the intima of human arteries and of its atherosclerosis-prone regions: a report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation.1992; 85:391-405. CrossrefMedlineGoogle Scholar
    • 156 Minick CR, Stemerman MG, Insull W. Effect of regenerated endothelium on lipid accumulation in the arterial wall. Proc Natl Acad Sci U S A.1977; 74:1724-1728. CrossrefMedlineGoogle Scholar
    • 157 Camejo G, Olofsson SO, Lopez F, Carlsson P, Bondjers G. Identification of apoB-100 segments mediating the interaction of low-density lipoproteins with arterial proteoglycans. Arteriosclerosis.1988; 8:368-377. LinkGoogle Scholar
    • 158 Ross R. The pathogenesis of atherosclerosis: an update. N Engl J Med.1986; 314:488-500. CrossrefMedlineGoogle Scholar
    • 159 Gordon D, Reidy MA, Benditt EP, Schwartz SM. Cell proliferation in human coronary arteries. Proc Natl Acad Sci U S A.1990; 87:4600-4604. CrossrefMedlineGoogle Scholar
    • 160 Ross R. Atherosclerosis: a defense mechanism gone awry. Am J Pathol.1993; 143:987-1002. MedlineGoogle Scholar
    • 161 Moss NS, Benditt EP. Human atherosclerotic plaque cells and leiomyoma cells: comparison of in vitro growth characteristics. Am J Pathol.1975; 78:175-190. MedlineGoogle Scholar
    • 162 Ross R, Wight TN, Strandness E, Thiele B. Human atherosclerosis, I: cell constitution and characteristics of advanced lesions of the superficial femoral artery. Am J Pathol.1984; 114:79-93. MedlineGoogle Scholar
    • 163 Dartsch PC, Voisard R, Bauriedel G, Hofling B, Betz E. Growth characteristics and cytoskeletal organization of cultured smooth muscle cells from human primary stenosing and restenosing lesions. Arteriosclerosis.1990; 10:62-75. LinkGoogle Scholar
    • 164 Bennett MR, Evan GI, Schwartz SM. Apoptosis of human vascular smooth muscle cells derived from normal vessels and coronary atherosclerotic plaques. J Clin Invest.1995; 95:2266-2274. CrossrefMedlineGoogle Scholar
    • 165 Hansson GK, Holm J, Holm S, Fotev Z, Hedrich HJ, Fingerle J. T lymphocytes inhibit the vascular response to injury. Proc Natl Acad Sci U S A.1991; 88:10530-10534. CrossrefMedlineGoogle Scholar
    • 166 O’Brien ER, Garvin MR, Dev R, Stewart DK, Hinohara T, Simpson JB, Schwartz SM. Angiogenesis in human coronary atherosclerotic plaques. Am J Pathol.1994; 145:883-894. MedlineGoogle Scholar
    • 167 Schwartz SM, Campbell GR, Campbell JH. Replication of smooth muscle cells in vascular disease. Circ Res.1986; 58:427-444. CrossrefMedlineGoogle Scholar
    • 168 Campbell GR, Campbell JH. The phenotypes of smooth muscle expressed in human atheroma. Ann N Y Acad Sci.1990; 598:143-158. CrossrefMedlineGoogle Scholar
    • 169 Desmouliere A, Gabbiani G. The cytoskeleton of arterial smooth muscle cells during human and experimental atheromatosis. Kidney Int. 1992;37(suppl):S87-S89. Google Scholar
    • 170 Hedin U, Sjolund M, Hultgardh-Nilsson A, Thyberg J. Changes in expression and organization of smooth muscle-specific α-actin during fibronectin-mediated modulation of arterial smooth muscle cell phenotype. Differentiation.1990; 44:222-231. CrossrefMedlineGoogle Scholar
    • 171 Thyberg J, Hedin U, Sjolund M, Palmberg L, Bottger BA. Regulation of differentiated properties and proliferation of arterial smooth muscle cells. Arteriosclerosis.1990; 10:966-990. LinkGoogle Scholar
    • 172 Kocher O, Gabbiani G. Cytoskeletal features of normal and atheromatous human arterial smooth muscle cells. Hum Pathol.1984; 17:875-880. CrossrefGoogle Scholar
    • 173 Frid MG, Moiseeva EP, Stenmark KR. Multiple phenotypically distinct smooth muscle cell populations exist in the adult and developing bovine pulmonary arterial media in vivo. Circ Res.1994; 75:669-681. CrossrefMedlineGoogle Scholar
    • 174 Clowes AW, Clowes MM, Kocher O, Ropraz P, Chaponnier C, Gabbiani G. Arterial smooth muscle cells in vivo: relationship between actin isoform expression and mitogenesis and their modulation by heparin. J Cell Biol.1988; 107:1939-1945. CrossrefMedlineGoogle Scholar
    • 175 Hood LC, Rosenquist TH. Coronary artery development in the chick: origin and deployment of smooth muscle cells, and the effects of neural crest ablation. Anat Rec.1992; 234:291-300. CrossrefMedlineGoogle Scholar
    • 176 Wight TN, Cooke PH, Smith SC. An electron microscopic study of pigeon aorta cell cultures: cytodifferentiation and intracellular lipid accumulation. Exp Mol Pathol.1977; 27:1-18. CrossrefMedlineGoogle Scholar
    • 177 Moss NS, Benditt EP. Spontaneous and experimentally induced arterial lesions, I: an ultrastructural survey of the normal chicken aorta. Lab Invest.1970; 22:166-183. MedlineGoogle Scholar
    • 178 Ludmer PL, Selwyn AP, Shook TL, Wayne RR, Mudge GL, Alexander RW, Ganz P. Paradoxical vasoconstriction induced by acetylcholine in atherosclerotic coronary arteries. N Engl J Med.1986; 315:1046-1051. CrossrefMedlineGoogle Scholar
    • 179 Flavahan NA. Atherosclerosis or lipoprotein-induced endothelial dysfunction: potential mechanisms underlying reduction in EDRF/nitric oxide activity. Circulation.1992; 85:1927-1938. LinkGoogle Scholar
    • 180 Joly GA, Schini VB, Vanhoutte PM. Balloon injury and interleukin-1β induce nitric oxide synthase activity in rat carotid arteries. Circ Res.1992; 71:331-338. CrossrefMedlineGoogle Scholar
    • 181 Winkles JA, Alberts GF, Brogi E, Libby P. Endothelin-1 and endothelin receptor mRNA expression in normal and atherosclerotic human arteries. Biochem Biophys Res Commun.1993; 191:1081-1088. CrossrefMedlineGoogle Scholar
    • 182 Glukhova M, Koteliansky V, Fondacci C, Marotte F, Rappaport L. Laminin variants and integrin laminin receptors in developing and adult human smooth muscle cells. Dev Biol.1993; 157:437-447. CrossrefMedlineGoogle Scholar
    • 183 Glukhova MA, Frid MG, Shekhonin BV, Vasilevskaya TD, Grunwald J, Saginati M, Koteliansky VE. Expression of extra domain A fibronectin sequence in vascular smooth muscle cells is phenotype-dependent. J Cell Biol.1989; 109:357-366. CrossrefMedlineGoogle Scholar
    • 184 Mackie EJ, Scott-Burden T, Hahn AW, Kern F, Bernhardt J, Regenass S, Weller A, Buhler FR. Expression of tenascin by vascular smooth muscle cells: alterations in hypertensive rats and stimulation by angiotensin II. Am J Pathol.1992; 141:377-388. MedlineGoogle Scholar
    • 185 Sharifi BG, LaFleur DW, Pirola CJ, Forrester JS, Fagin JA. Angiotensin II regulates tenascin gene expression in vascular smooth muscle cells. J Biol Chem.1992; 267:23910-23915. CrossrefMedlineGoogle Scholar
    • 186 Niimi Y, Azuma H, Hirakawa K. Repeated endothelial removal augments intimal thickening and attenuates EDRF release. Am J Physiol.1994; 266:H1348-H1356. MedlineGoogle Scholar
    • 187 Majesky MW, Giachelli CM, Reidy MA, Schwartz SM. Rat carotid neointimal smooth muscle cells reexpress a developmentally regulated mRNA phenotype during repair of arterial injury. Circ Res.1992; 71:759-768. CrossrefMedlineGoogle Scholar
    • 188 Schwartz SM, Foy L, Bowen-Pope DF, Ross R. Derivation and properties of platelet-derived growth factor-independent rat smooth muscle cells. Am J Pathol.1990; 136:1417-1428. MedlineGoogle Scholar
    • 189 Giachelli CM, Majesky MW, Schwartz SM. Developmentally regulated cytochrome P450IA1 expression in cultured rat vascular smooth muscle cells. J Biol Chem.1991; 266:3981-3986. CrossrefMedlineGoogle Scholar
    • 190 Giachelli CM, Bae N, Lombardi DM, Majesky MW, Schwartz SM. Molecular cloning and characterization of 2B7, a rat mRNA which distinguishes smooth muscle cell phenotypes in vitro and is identical to osteopontin (secreted phosphoprotein I, 2aR). Biochem Biophys Res Commun.1991; 177:867-873. CrossrefMedlineGoogle Scholar
    • 191 Campbell GR, Campbell JH. Smooth muscle phenotypic changes in arterial wall homeostasis: implications for the pathogenesis of atherosclerosis. Exp Mol Pathol.1984; 42:139-162. Google Scholar
    • 192 Lemire JM, Covin CW, White S, Giachelli CM, Schwartz SM. Characterization of cloned aortic smooth muscle cells from young rats. Am J Pathol.1994; 144:1068-1081. MedlineGoogle Scholar
    • 193 Steinberg D. Antioxidant vitamins and coronary heart disease. N Engl J Med.1993; 328:1444-1449. CrossrefMedlineGoogle Scholar
    • 194 Libby P, Hansson GK. Involvement of the immune system in human atherogenesis: current knowledge and unanswered questions. Lab Invest.1991; 64:5-15. MedlineGoogle Scholar
    • 195 Collins T. Endothelial nuclear factor-κB and the initiation of the atherosclerotic lesion. Lab Invest.1993; 68:499-508. MedlineGoogle Scholar
    • 196 Davies MJ, Thomas AC. Plaque fissuring: the cause of acute myocardial infarction, sudden ischemic death, and crescendo angina. Br Heart J.1985; 53:363-373. CrossrefMedlineGoogle Scholar
    • 197 Wilcox JN, Smith KM, Schwartz SM, Gordon D. Localization of tissue factor in the normal vessel wall and in the atherosclerotic plaque. Proc Natl Acad Sci U S A.1989; 86:2839-2843. CrossrefMedlineGoogle Scholar
    • 198 Edgington TS, Mackman N, Brand K, Ruf W. The structural biology of expression and function of tissue factor. Thromb Haemost.1991; 66:67-79. CrossrefMedlineGoogle Scholar
    • 199 Constantinides P. Plaque fissures in human coronary thrombosis. J Atheroscler Res.1966; 6:1-17. CrossrefGoogle Scholar
    • 200 Falk E. Morphologic features of unstable atherothrombotic plaques underlying acute coronary syndromes. Am J Cardiol.1989; 63:114E-120E. CrossrefMedlineGoogle Scholar
    • 201 Taubman MB. Tissue factor regulation in vascular smooth muscle: a summary of studies performed using in vivo and in vitro models. Am J Cardiol.1993; 72:55C-60C. CrossrefMedlineGoogle Scholar
    • 202 Giachelli CM, Bae N, Almeida M, Denhardt DT, Alpers CE, Schwartz SM. Osteopontin is elevated during neointima formation in rat arteries and is a novel component of human atherosclerotic plaques. J Clin Invest.1993; 92:1686-1696. CrossrefMedlineGoogle Scholar
    • 203 Bostrom K, Watson KE, Horn S, Wortham C, Herman IM, Demer LL. Bone morphogenetic protein expression in human atherosclerotic lesions. J Clin Invest.1993; 91:1800-1809. CrossrefMedlineGoogle Scholar
    • 204 Glagov S, Weisenberg E, Zarins CK, Stankunavicius R, Kolettis GJ. Compensatory enlargement of human atherosclerotic coronary arteries. N Engl J Med.1987; 316:1371-1375. CrossrefMedlineGoogle Scholar
    • 205 Folkow B. Physiological aspects of primary hypertension. Physiol Rev.1982; 62:347-504. CrossrefMedlineGoogle Scholar
    • 206 Jamal A, Bendeck M, Langille BL. Structural changes and recovery of function after arterial injury. Arterioscler Thromb.1992; 12:307-317. CrossrefMedlineGoogle Scholar
    • 207 Dilley RJ, Schwartz SM. Vascular remodeling in the growth hormone transgenic mouse. Circ Res.1989; 65:1233-1240. CrossrefMedlineGoogle Scholar
    • 208 Mulvany MJ. Determinants of vascular structure. J Cardiovasc Pharmacol. 1992;19(suppl 5):S1-S6. Google Scholar
    • 209 Koester W. Endarteritis and arteritis. Berl Klin Wochenschr.1876; 13:454-455. Google Scholar
    • 210 Geiringer E. Intimal vascularization and atherosclerosis. J Pathol Bacteriol.1951; 63:201-211. CrossrefMedlineGoogle Scholar
    • 211 Barger AC, Beeuwkes RI, Lainey LL, Silverman KJ. Vasa vasorum and neovascularization of human coronary arteries. N Engl J Med.1984; 310:175-177. CrossrefMedlineGoogle Scholar
    • 212 Brown BG, Albers JJ, Fisher LD, Schaefer SM, Lin JT, Kaplan CK, Zhao XQ, Bisson BD, Fitzpatrick VF, Dodge HT. Regression of coronary artery disease as a result of intensive lipid-lowering therapy in men with high levels of apolipoprotein B. N Engl J Med.1990; 323:1289-1298. CrossrefMedlineGoogle Scholar
    • 213 Blankenhorn DH, Nessim SA, Johnson RL, Sanmarco RE, Azen SP, Cachin-Hamphill L. Beneficial effects of colestipol niacin therapy on coronary atherosclerosis and coronary venous bypass grafts. JAMA.1987; 257:3233-3240. CrossrefMedlineGoogle Scholar
    • 214 Watts GF, Lewis B, Brunt JN, Lewis ES, Coltart DJ, Smith LD, Mann JI, Swan AV. Effects on coronary artery disease of lipid-lowering diet, or diet plus cholestyramine, in the St. Thomas’ Atherosclerosis Regression Study (STARS). Lancet.1992; 339:563-569. CrossrefMedlineGoogle Scholar
    • 215 Gould KL. Quantitative analysis of coronary artery restenosis after coronary angioplasty: Has the rose lost its bloom? J Am Coll Cardiol.1992; 19:946-947. CrossrefMedlineGoogle Scholar
    • 216 Kragel AH, Reddy SG, Wittes JT, Roberts WC. Morphometric analysis of the composition of atherosclerotic plaques in the four major epicardial coronary arteries in acute myocardial infarction and in sudden coronary death. Circulation.1989; 80:1747-1765. CrossrefMedlineGoogle Scholar
    • 217 Scandinavian Simvastatin Survival Group Study. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). Lancet.1994; 344:1383-1389. MedlineGoogle Scholar
    • 218 Henney AM, Wakeley PR, Davies MJ, Foster K, Hembry R, Murphy G, Humphries S. Localization of stromelysin gene expression in atherosclerotic plaques by in situ hybridization. Proc Natl Acad Sci U S A.1991; 88:8154-8158. CrossrefMedlineGoogle Scholar
    • 219 Davies MJ. Anatomic features in victims of sudden coronary death: coronary artery pathology. Circulation. 1992;85(suppl I):I-19-I-24. Google Scholar
    • 220 Prescott MF, McBride CH, Hasler-Rapacz J, Von Linden J, Rapacz J. Development of complex atherosclerotic lesions in pigs with inherited hyper-LDL cholesterolemia bearing mutant alleles for apolipoprotein B. Am J Pathol.1991; 139:139-147. MedlineGoogle Scholar
    • 221 Little WC, Constantinescu M, Applegate RJ, Kutcher MA, Burrows MT, Kahl FR, Santamore WP. Can coronary angiography predict the site of a subsequent myocardial infarction in patients with mild-to-moderate coronary artery disease? Circulation. 1988;78(pt 1):1157-1166. Google Scholar
    • 222 Ambrose JA, Tannenbaum MA, Alexopoulos D, Hjemdahl-Monsen CE, Leavy J, Weiss M, Borrico S, Gorlin R, Fuster V. Angiographic progression of coronary artery disease and the development of myocardial infarction. J Am Coll Cardiol.1988; 12:56-62. CrossrefMedlineGoogle Scholar
    • 223 Qiao JH, Walts AE, Fishbein MC. The severity of atherosclerosis at sites of plaque rupture with occlusive thrombosis in saphenous vein coronary artery bypass grafts. Am Heart J. 1991;122(pt 1):955-958. Google Scholar
    • 224 Qiao JH, Fishbein MC. The severity of coronary atherosclerosis at sites of plaque rupture with occlusive thrombosis. J Am Coll Cardiol.1991; 17:1138-1142. CrossrefMedlineGoogle Scholar
    • 225 Nobuyoshi M, Kimura T, Ohishi H, Horiuchi H, Nosaka H, Hamasaki N, Yokoi H, Kim K. Restenosis after percutaneous transluminal coronary angioplasty: pathologic observations in 20 patients. J Am Coll Cardiol.1991; 17:433-439. CrossrefMedlineGoogle Scholar
    • 226 McCormick D, Hall PA. The complexities of proliferating cell nuclear antigen. Histopathology.1992; 21:591-594. CrossrefMedlineGoogle Scholar
    • 227 O’Brien ER, Alpers CE, Stewart DK, Ferguson M, Tran N, Gordon D, Benditt EP, Hinohara T, Simpson JB, Schwartz SM. Proliferation in primary and restenotic coronary atherectomy tissue: implications for antiproliferative therapy. Circ Res.1993; 73:223-231. CrossrefMedlineGoogle Scholar
    • 228 Leclerc G, Kearney M, Schneider D, Rosenfield K, Losordo DW, Isner JM. Assessment of cell kinetics in human restenotic lesions by in vitro bromodeoxyuridine labeling of excised atherectomy specimens. Clin Res.1993; 41:343A. Abstract. Google Scholar
    • 229 Strauss BH, Umans VA, VanSuylen RJ, deFeyter PJ, Marco J, Robertson GC, Renkin J, Heyndrickx G, Vuzevski VD, Bosman FT. Directional atherectomy for treatment of restenosis within coronary stents: clinical, angiographic and histological results. J Am Coll Cardiol.1992; 20:1465-1473. CrossrefMedlineGoogle Scholar
    • 230 Rekhter M, Nicholls S, Ferguson M, Gordon D. Cell proliferation in human arteriovenous fistulas used for hemodialysis. Arterioscler Thromb.1993; 13:609-617. CrossrefMedlineGoogle Scholar
    • 231 Pickering JG, Weir L, Jekanowski J, Kearney MA, Isner JM. Proliferative activity in peripheral and coronary atherosclerotic plaque among patients undergoing percutaneous revascularization. J Clin Invest.1993; 91:1469-1480. CrossrefMedlineGoogle Scholar
    • 232 Garcia RL, Coltrera MD, Gown AM. Analysis of proliferative grade using anti-PCNA/cyclin monoclonal antibodies in fixed, embedded tissues: comparison with flow cytometric analysis. Am J Pathol.1989; 134:733-739. MedlineGoogle Scholar
    • 233 Miller MJ, Kuntz RE, Friedrich SP, Leidig GA, Fishman RF, Schnitt SJ, Baim DS, Safian RD. Frequency and consequences of intimal hyperplasia in specimens retrieved by directional atherectomy of native primary coronary artery stenoses and subsequent restenoses. Am J Cardiol.1993; 71:652-658. CrossrefMedlineGoogle Scholar
    • 234 Schnitt SJ, Safian RD, Kuntz RE, Schmidt DA, Baim DS. Histologic findings in specimens obtained by percutaneous directional coronary atherectomy. Hum Pathol.1992; 23:415-420. CrossrefMedlineGoogle Scholar
    • 235 Orekhov AN, Ankarpova II, Tertov VV, Rudchenko SA, Addreeva ER, Krushinsky AV, Smirnov RN. Cellular composition of atherosclerotic and uninvolved human aortic subendothelial intima: light-microscopic study of dissociated aortic cells. Am J Pathol.1984; 115:17-24. MedlineGoogle Scholar
    • 236 Andreeva ER, Rekhter MD, Romanov YA, Antonova GM, Antonov AS, Mironov AA, Orekhov AN. Stellate cells of aortic intima, II: arborization of intimal cells in culture. Tissue Cell.1992; 24:697-704. CrossrefMedlineGoogle Scholar
    • 237 Lafont AM, Chisolm GM, Whitlow PL, Goormastic M, Cornhill JF. Post-angioplasty restenosis in the atherosclerotic rabbit: proliferative response or chronic constriction? Circulation. 1993;88(pt 2):I-521. Abstract. Google Scholar
    • 238 Kakuta T, Currier JW, Haudenschild CC, Ryan TJ, Faxon DP. Differences in compensatory vessel enlargement, not intimal formation, account for restenosis after angioplasty in the hypercholesterolemic rabbit model. Circulation.1994; 89:2809-2815. CrossrefMedlineGoogle Scholar
    • 239 Nunes GL, Sigman SR, Robinson KA, Cipolla GD, Gravanis MB, Berk BC. Improved quantification of changes in vessel morphology after balloon injury using linear regression analysis. Circulation. 1993;88(suppl I, pt 2):I-650. Abstract. Google Scholar
    • 240 Brott BC, Labinaz M, Culp SC, Zidar JP, Fortin DF, Virmani R, Philips HR, Stack RS. Vessel remodeling after angioplasty: comparative anatomic studies. J Am Coll Cardiol.1994; 23:138A. Abstract. Google Scholar
    • 241 Langille BL, O’Donnell F. Reductions in arterial diameter produced by chronic diseases in blood flow are endothelium-dependent. Science.1986; 231:405-407. CrossrefMedlineGoogle Scholar
    • 242 Post MJ, Borst C, Kuntz RE. The relative importance of arterial remodeling compared with intimal hyperplasia in lumen renarrowing after balloon angioplasty: a study in the normal rabbit and the hypercholesterolemic Yucatan micropig. Circulation.1994; 89:2816-2821. CrossrefMedlineGoogle Scholar
    • 243 Dotter CT, Judkins MP. Transluminal treatment of atherosclerotic obstructions: description of new technique and a preliminary report of its application. Circulation.1964; 30:654-670. CrossrefMedlineGoogle Scholar
    • 244 Gruentzig AR, Myler RK, Hanna EH, Turina MI. Restenosis after balloon angioplasty. Circulation. 1977;84(suppl II):II-55-II-56. Google Scholar
    • 245 Lee G, Ikeda RM, Joye JA, Bogren HG, Demaria AN, Mason DT. Evaluation of transluminal angioplasty of chronic coronary artery stenosis. Circulation.1980; 61:77-83. CrossrefMedlineGoogle Scholar
    • 246 Block PC, Myler RK, Stertzer S, Fallon JT. Morphology after transluminal angioplasty in human beings. N Engl J Med.1981; 305:382-385. CrossrefMedlineGoogle Scholar
    • 247 Castaneda-Zuniga WR, Formanek A, Tadavarthy M, Vlodaver Z, Edwards JE, Zollikofer C, Amplatz K. The mechanism of balloon angioplasty. Radiology.1980; 135:565-571. CrossrefMedlineGoogle Scholar
    • 248 Baughman KL, Pasternak RC, Fallon JT, Block PC. Transluminal coronary angioplasty of postmortem human hearts. Am J Cardiol.1981; 48:1044-1047. CrossrefMedlineGoogle Scholar
    • 249 Waller BF. Pathology of transluminal balloon angioplasty used in the treatment of coronary heart disease. Hum Pathol.1987; 18:476-484. CrossrefMedlineGoogle Scholar
    • 250 Mizuno K, Jurita A, Imazeki N. Pathologic findings after percutaneous transluminal coronary angioplasty. Br Heart J.1984; 52:588-590. CrossrefMedlineGoogle Scholar
    • 251 Losordo DW, Rosenfield K, Pieczek A, Baker K, Harding M, Isner JM. How does angioplasty work? Serial analysis of human iliac arteries using intravascular ultrasound. Circulation.1992; 86:1845-1858. CrossrefMedlineGoogle Scholar
    • 252 Mintz GS, Douek PC, Bonner RF, Kent KM, Pichard AD, Satler LF, Leon MB. Intravascular ultrasound comparison of de novo and restenotic coronary artery lesions. J Am Coll Cardiol.1993; 21:118A. Abstract. Google Scholar
    • 253 Mintz GS, Kovach JA, Javier SP, Ditrano CJ, Leon MB. Geometric remodeling is the predominant mechanism of late lumen loss after coronary angioplasty. Circulation. 1993;88(suppl I, pt 2):I-654. Abstract. Google Scholar
    • 254 Schwartz RS, Koval TM, Edwards WD, Camrud AR, Bailey KR, Browne K, Vlietstra RE, Holmes DR Jr. Effect of external beam irradiation on neointimal hyperplasia after experimental coronary artery injury. J Am Coll Cardiol.1992; 19:1106-1113. CrossrefMedlineGoogle Scholar
    • 255 Chang MY, Sasahara M, Raines EW, Chait A, Ross R. PCNA-positive cells are decreased in lesions by probucol treatment in hypercholesterolemic nonhuman primates. Circulation. 1993;88(suppl I, pt 2):I-564. Abstract. Google Scholar
    • 256 Lawn RM, Wade DP, Hammer RE, Chiesa G, Verstuyft JG, Rubin EM. Atherogenesis in transgenic mice expressing human apolipoprotein(a). Nature.1992; 360:670-672. CrossrefMedlineGoogle Scholar
    • 257 Lovastatin restenosis trial study group. Lovastatin restenosis trial: results in patients with cholesterol over 200 mg/dl. J Am Coll Cardiol.1994; 23:470A. Abstract. CrossrefMedlineGoogle Scholar
    • 258 Sawa H, Sobel BE, Fujii S. Potentiation by hypercholesterolemia of the induction of aortic intramural synthesis of plasminogen activator inhibitor type 1 by endothelial injury. Circ Res.1993; 73:671-680. CrossrefMedlineGoogle Scholar
    • 259 Glukhova MA, Frid MG, Koteliansky VE. Phenotypic changes of human aortic smooth muscle cells during development and in the adult vessel. Am J Physiol. 1991;261(suppl):78-80. Google Scholar
    • 260 Yla-Herttuala S. Gene expression in atherosclerotic lesions. Herz.1992; 17:270-276. MedlineGoogle Scholar
    • 261 Dzau VJ. Local expression and pathophysiological role of renin-angiotensin in the blood vessels and heart. Basic Res Cardiol. 1993;88(suppl 1):1-14. Google Scholar
    • 262 Tipping PG, Davenport P, Gallicchio M, Filonzi EL, Apostolopoulos J, Wojta J. Atheromatous plaque macrophages produce plasminogen activator inhibitor type-1 and stimulate its production by endothelial cells and vascular smooth muscle cells. Am J Pathol.1993; 143:875-885. MedlineGoogle Scholar
    • 263 Schneiderman J, Sawdey MS, Keeton MR, Bordin GM, Bernstein EF, Dilley RB, Loskutoff DJ. Increased type 1 plasminogen activator inhibitor gene expression in atherosclerotic human arteries. Proc Natl Acad Sci U S A.1992; 89:6998-7002. CrossrefMedlineGoogle Scholar
    • 264 Lupu F, Bergonzelli GE, Heim DA, Cousin E, Genton CY, Bachmann F, Kruithof EK. Localization and production of plasminogen activator inhibitor-1 in human healthy and atherosclerotic arteries. Arterioscler Thromb.1993; 13:1090-1100. CrossrefMedlineGoogle Scholar
    • 265 Fink LM, Eidt JF, Johnson K, Cook JM, Cook CD, Morser J, Marlar R, Collins CL, Schaefer R, Xie SS. Thrombomodulin activity and localization. Int J Dev Biol.1993; 37:221-226. MedlineGoogle Scholar
    • 266 Kocher O, Skalli O, Bloom WS, Gabbiani G. Cytoskeleton of rat aortic smooth muscle cells: normal conditions and experimental intimal thickening. Lab Invest.1984; 50:645-652. MedlineGoogle Scholar
    • 267 Shanahan CM, Weissberg PL, Metcalfe JC. Isolation of gene markers of differentiated and proliferating vascular smooth muscle cells. Circ Res.1993; 73:193-204. CrossrefMedlineGoogle Scholar
    • 268 Hansson GK, Hellstrand M, Rymo L, Rubbia L, Gabbiani G. Interferon gamma inhibits both proliferation and expression of differentiation-specific α-smooth muscle actin in arterial smooth muscle cells. J Exp Med.1989; 170:1595-1608. CrossrefMedlineGoogle Scholar
    • 269 Glukhova MA, Frid MG, Koteliansky VE. Developmental changes in expression of contractile and cytoskeletal proteins in human aortic smooth muscle. J Biol Chem.1990; 265:13042-13046. CrossrefMedlineGoogle Scholar
    • 270 Bochaton-Piallat ML, Gabbiani F, Ropraz P, Gabbiani G. Cultured aortic smooth muscle cells from newborn and adult rats show distinct cytoskeletal features. Differentiation.1992; 49:175-185. CrossrefMedlineGoogle Scholar
    • 271 Frid MG, Shekhonin BV, Koteliansky VE, Glukhova MA. Phenotypic changes of human smooth muscle cells during development: late expression of heavy caldesmon and calponin. Dev Biol.1992; 153:185-193. CrossrefMedlineGoogle Scholar
    • 272 Jahn L, Kreuzer J, von Hodenberg E, Kubler W, Franke WW, Allenberg J, Izumo S. Cytokeratins 8 and 18 in smooth muscle cells: detection in human coronary artery, peripheral vascular, and vein graft disease and in transplantation-associated arteriosclerosis. Arterioscler Thromb.1993; 13:1631-1639. CrossrefMedlineGoogle Scholar
    • 273 Babaev VR, Bobryshev YV, Stenina OV, Tararak EM, Gabbiani G. Heterogeneity of smooth muscle cells in atheromatous plaque of human aorta. Am J Pathol.1990; 136:1031-1042. MedlineGoogle Scholar
    • 274 Birukov KG, Frid MG, Rogers JD, Shirinsky VP. Synthesis and expression of smooth muscle phenotype markers in primary culture of rabbit aortic smooth muscle cells: influence of seeding density and media and relation to cell contractility. Exp Cell Res.1993; 204:46-53. CrossrefMedlineGoogle Scholar
    • 275 Giuriato L, Scatena M, Chiavegato A, Tonello M, Scannapieco G, Pauletto P, Sartore S. Non-muscle myosin isoforms and cell heterogeneity in developing rabbit vascular smooth muscle. J Cell Sci.1992; 101:233-246. CrossrefMedlineGoogle Scholar
    • 276 Grainger DJ, Hesketh TR, Metcalfe JC, Weissberg PL. A large accumulation of non-muscle myosin occurs at first entry into M phase in rat vascular smooth muscle cells. Biochem J.1991; 277:145-151. CrossrefMedlineGoogle Scholar
    • 277 Simons M, Leclerc G, Safian RD, Isner JM, Weir L, Baim DS. Relation between activated smooth muscle cells in coronary artery lesions and restenosis after atherectomy. N Engl J Med.1993; 328:608-613. CrossrefMedlineGoogle Scholar
    • 278 Leclerc G, Isner JM, Kearney M, Simons M, Safian RD, Baim DS, Weir L. Evidence implicating nonmuscle myosin in restenosis: use of in situ hybridization to analyze human vascular lesions obtained by directional atherectomy. Circulation.1992; 85:543-553. CrossrefMedlineGoogle Scholar
    • 279 Chaponnier C, Kocher O, Gabbiani G. Modulation of gelsolin content in rat aortic smooth muscle cells during development, experimental intimal thickening and culture: an immunohistochemical and biochemical study. Eur J Biochem.1990; 190:559-565. CrossrefMedlineGoogle Scholar
    • 280 Pauletto P, Sartore S, Giuriato L, Scatena M, Tonello M, Scannapieco G, Pessina AC, Dal-Palu C. Calcium antagonists and vascular smooth muscle cells in atherogenesis. J Cardiovasc Pharmacol. 1992;19(suppl 2):S8-S16. Google Scholar
    • 281 Babaev VR, Antonov AS, Domogatsky SP, Kazantseva IA. Phenotype related changes of intimal smooth muscle cells from human aorta in primary culture. Atherosclerosis.1992; 96:189-202. CrossrefMedlineGoogle Scholar
    • 282 Grainger DJ, Hesketh TR, Weissberg PL, Metcalfe JC. Hexamethylenebisacetamide selectively inhibits the proliferation of human and rat vascular smooth muscle cells. Biochem J.1992; 283:403-408. CrossrefMedlineGoogle Scholar
    • 283 Giuriato L, Scatena M, Chiavegato A, Zanellato AM, Guidolin D, Pauletto P, Sartore S. Localization and smooth muscle cell composition of atherosclerotic lesions in Watanabe heritable hyperlipidemic rabbits. Arterioscler Thromb.1993; 13:347-359. CrossrefMedlineGoogle Scholar
    • 284 Zanellato AM, Borrione AC, Tonello M, Scannapieco G, Pauletto P, Sartore S. Myosin isoform expression and smooth muscle cell heterogeneity in normal and atherosclerotic rabbit aorta. Arteriosclerosis.1990; 10:996-1009. LinkGoogle Scholar
    • 285 Zanellato AM, Borrione AC, Giuriato L, Tonello M, Scannapieco G, Pauletto P, Sartore S. Myosin isoforms and cell heterogeneity in vascular smooth muscle, I: developing and adult bovine aorta. Dev Biol.1990; 141:431-446. CrossrefMedlineGoogle Scholar
    • 286 Patel CV, Walsh K. Regulation of major histocompatibility gene expression in human vascular smooth muscle cells. Arteriosclerosis.1989; 9:279-288. LinkGoogle Scholar
    • 287 Liau G, Chan LM, Feng P. Increased ferritin gene expression is both promoted by cAMP and a marker of growth arrest in rabbit vascular smooth muscle cells. J Biol Chem.1991; 266:18819-18826. CrossrefMedlineGoogle Scholar
    • 288 Speir E, Sasse J, Shrivastav S, Casscells W. Culture-induced increase in acidic and basic fibroblast growth factor activities and their association with the nuclei of vascular endothelial and smooth muscle cells. J Cell Physiol.1991; 147:362-373. CrossrefMedlineGoogle Scholar
    • 289 Reidy MA, Lindner V. Basic FGF and growth of arterial cells.Ann N Y Acad Sci.1991; 638:290-299. CrossrefMedlineGoogle Scholar
    • 290 Khorsandi MJ, Fagin JA, Giannella-Neto D, Forrester JS, Cercek B. Regulation of insulin-like growth factor-I and its receptor in rat aorta after balloon denudation: evidence for local bioactivity. J Clin Invest.1992; 90:1926-1931. CrossrefMedlineGoogle Scholar
    • 291 Giannella-Neto D, Kamyar A, Sharifi B, Pirola CJ, Kupfer J, Rosenfeld RG, Forrester JS, Fagin JA. Platelet-derived growth factor isoforms decrease insulin-like growth factor I gene expression in rat vascular smooth muscle cells and selectively stimulate the biosynthesis of insulin-like growth factor binding protein 4. Circ Res.1992; 71:646-656. CrossrefMedlineGoogle Scholar
    • 292 Khorsandi M, Fagin JA, Fishbein MC, Forrester JS, Cercek B. Effects of hypophysectomy on vascular insulin-like growth factor-I gene expression after balloon denudation in rats. Atherosclerosis.1992; 93:115-122. CrossrefMedlineGoogle Scholar
    • 293 Han DKM, Liau G. Identification and characterization of developmentally regulated genes in vascular smooth muscle cells. Circ Res.1992; 71:711-719. CrossrefMedlineGoogle Scholar
    • 294 Okazaki H, Majesky MW, Harker LA, Schwartz SM. Regulation of platelet-derived growth factor ligand and receptor gene expression by α-thrombin in vascular smooth muscle cells. Circ Res.1992; 71:1285-1293. CrossrefMedlineGoogle Scholar
    • 295 Warner SJ, Friedman GB, Libby P. Regulation of major histocompatibility gene expression in human vascular smooth muscle cells. Arteriosclerosis.1989; 9:279-288. LinkGoogle Scholar
    • 296 Jonasson L, Holm J, Skalli O, Gabbiani G, Hansson GK. Expression of class II transplantation antigen on vascular smooth muscle cells in human atherosclerosis. J Clin Invest.1985; 76:125-131. CrossrefMedlineGoogle Scholar
    • 297 Poston RN, Haskard DO, Coucher JR, Gall NP, Johnson-Tidey RR. Expression of intercellular adhesion molecule-1 in atherosclerotic plaques. Am J Pathol.1992; 140:665-673. MedlineGoogle Scholar
    • 298 Wawryk SO, Ayberk H, Boyd AW, Rode J. Analysis of adhesion molecules in the immunopathogenesis of giant cell arteries. J Clin Pathol.1991; 44:497-501. CrossrefMedlineGoogle Scholar
    • 299 Loppnow H, Libby P. Proliferating or interleukin 1-activated human vascular smooth muscle cells secrete copious interleukin 6. J Clin Invest.1990; 85:731-738. CrossrefMedlineGoogle Scholar
    • 300 Moyer CF, Sajuthi D, Tulli H, Williams JK. Synthesis of IL-1α and IL-1β by arterial cells in atherosclerosis. Am J Pathol.1991; 138:951-960. MedlineGoogle Scholar
    • 301 Clinton SK, Fleet JC, Loppnow H, Salomon RN, Clark BD, Cannon JG, Shaw AR, Dinarello CA, Libby P. Interleukin-1 gene expression in rabbit vascular tissue in vivo. Am J Pathol.1991; 138:1005-1014. MedlineGoogle Scholar
    • 302 Arbustini E, Grasso M, Diegoli M, Pucci A, Bramerio M, Ardissino D, Angoli L, de Servi S, Bramucci E, Mussini A. Coronary atherosclerotic plaques with and without thrombus in ischemic heart syndromes: a morphologic, immunohistochemical, and biochemical study. Am J Cardiol.1991; 68:36B-50B. CrossrefMedlineGoogle Scholar
    • 303 Koch AE, Kunkel SL, Pearce WH, Shah MR, Parikh D, Evanoff HL, Haines GK, Burdick MD, Strieter RM. Enhanced production of the chemotactic cytokines interleukin-8 and monocyte chemoattractant protein-1 in human abdominal aortic aneurysms. Am J Pathol.1993; 142:1423-1431. MedlineGoogle Scholar
    • 304 Wang JM, Sica A, Peri G, Walter S, Padura IM, Libby P, Ceska M, Lindley I, Colotta F, Mantovani A. Expression of monocyte chemotactic protein and interleukin-8 by cytokine-activated human vascular smooth muscle cells. Arterioscler Thromb.1991; 11:1166-1174. CrossrefMedlineGoogle Scholar
    • 305 Clinton SK, Underwood R, Hayes L, Sherman ML, Kufe DW, Libby P. Macrophage colony-stimulating factor gene expression in vascular cells and in experimental and human atherosclerosis. Am J Pathol.1992; 140:301-316. MedlineGoogle Scholar
    • 306 Rus HG, Niculescu F, Vlaicu R. Tumor necrosis factor-α in human atherosclerosis. Atherosclerosis.1991; 89:247-254. CrossrefMedlineGoogle Scholar
    • 307 Barath P, Fishbein MC, Cao J, Berenson J, Helfant RH, Forrester JS. Tumor necrosis factor gene expression in human vascular intimal smooth muscle cells detected by in situ hybridization. Am J Pathol.1990; 137:503-509. MedlineGoogle Scholar
    • 308 O’Brien KD, Allen MD, McDonald TO, Chait A, Harlan JM, Fishbein D, McCarty J, Ferguson M, Hudkins K, Benjamin CD. Vascular cell adhesion molecule-1 is expressed in human coronary atherosclerotic plaques: implications for the mode of progression of advanced coronary atherosclerosis. J Clin Invest.1993; 92:945-951. CrossrefMedlineGoogle Scholar
    • 309 Briscoe DM, Cotran RS, Pober JS. Effects of tumor necrosis factor, lipopolysaccharide, and IL-4 on the expression of vascular cell adhesion molecule-1 in vivo: correlation with CD3+ T cell infiltration. J Immunol.1992; 149:2954-2960. CrossrefMedlineGoogle Scholar
    • 310 Inaba T, Gotoda T, Shimano H, Shimada M, Harada K, Kozaki K, Watanabe Y, Hoh E, Motoyoshi K, Yazaki Y. Platelet-derived growth factor induces c-fms and scavenger receptor genes in vascular smooth muscle cells. J Biol Chem.1992; 267:13107-13112. CrossrefMedlineGoogle Scholar
    • 311 Bickel PE, Freeman MW. Rabbit aortic smooth muscle cells express inducible macrophage scavenger receptor messenger RNA that is absent from endothelial cells. J Clin Invest.1992; 90:1450-1457. CrossrefMedlineGoogle Scholar
    • 312 Salomon RN, Underwood R, Doyle MV, Wang A, Libby P. Increased apolipoprotein E and c-fms gene expression without elevated interleukin 1 or 6 mRNA levels indicates selective activation of macrophage functions in advanced human atheroma. Proc Natl Acad Sci U S A.1992; 89:2814-2818. CrossrefMedlineGoogle Scholar
    • 313 Jonasson L, Bondjers G, Hansson GK. Lipoprotein lipase in atherosclerosis: its presence in smooth muscle cells and absence from macrophages. J Lipid Res.1987; 28:437-445. CrossrefMedlineGoogle Scholar
    • 314 Yla-Herttuala S, Lipton BA, Rosenfeld ME, Sarkioja T, Yoshimura T, Leonard EJ, Witztum JL, Steinberg D. Expression of monocyte chemoattractant protein 1 in macrophage-rich areas of human and rabbit atherosclerotic lesions. Proc Natl Acad Sci U S A.1991; 88:5252-5256. CrossrefMedlineGoogle Scholar
    • 315 Navab M, Imes SS, Hama SY, Hough GP, Ross LA, Bork RW, Valente AJ, Berliner JA, Drinkwater DC, Laks H. Monocyte transmigration induced by modification of low density lipoprotein in cocultures of human aortic wall cells is due to induction of monocyte chemotactic protein 1 synthesis and is abolished by high density lipoprotein. J Clin Invest.1991; 88:2039-2046. CrossrefMedlineGoogle Scholar
    • 316 Gmi’nski J, Dr’ozdz M, Ulfig-Ma’slanka R, Najda J. Evaluation of elastin metabolism in children from families with high risk of atherosclerosis. Atherosclerosis.1991; 91:185-189. CrossrefMedlineGoogle Scholar
    • 317 Stone PJ, McMahon MP, Morris SM, Calore JD, Franzblau C. Elastin in a neonatal rat smooth muscle cell culture has greatly decreased susceptibility to proteolysis by human neutrophil elastase: an in vitro model of elastolytic injury. In Vitro Cell Dev Biol.1987; 23:663-676. CrossrefMedlineGoogle Scholar
    • 318 Gijsbers BL, van Haarlem LJ, Soute BA, Ebberink RH, Vermeer C. Characterization of a Gla-containing protein from calcified human atherosclerotic plaques. Arteriosclerosis.1990; 10:991-995. LinkGoogle Scholar
    • 319 Hedin U, Holm H, Hansson G. Induction of tenascin in rat arterial injury: relationship to altered smooth muscle cell phenotype. Am J Pathol.1991; 139:649-656. MedlineGoogle Scholar
    • 320 Lark MW, Yeo TK, Mar H, Lara S, Hellstrom I, Hellstrom KE, Wight TN. Arterial chondroitin sulfate proteoglycan: localization with a monoclonal antibody. J Histochem Cytochem.1988; 36:1211-1221. CrossrefMedlineGoogle Scholar
    • 321 Smith EB, Staples EM. Haemostatic factors in human aortic intima. Lancet.1981; 1:1171-1174. CrossrefMedlineGoogle Scholar
    • 322 Wilcox JN. Thrombin and other potential mechanisms underlying restenosis. Circulation.1991; 84:432-435. CrossrefMedlineGoogle Scholar

    eLetters(0)

    eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

    Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.