Literature
首页医源资料库在线期刊动脉硬化血栓血管生物学杂志2006年第26卷第6期

Modulation of Smooth Muscle Cell Migration by Members of the Low-Density Lipoprotein Receptor Family

来源:《动脉硬化血栓血管生物学杂志》
摘要:21ModulationoftheLR11/uPARPathwayforPreventionofAtherosclerosisStatinsarepotentinhibitorsofthe3-hydroxy-3-methylglutaryl-coenzymeAreductasethatareknowntobeeffectiveforpreventingatherosclerosis。JCellSci。Cell。JCellBiol。...

点击显示 收起

【摘要】  Low-density lipoprotein receptor family members (LRs) play a key role in the catabolism of many membrane-associated proteins, such as complexes between proteinases and their receptors, in addition to being involved in lipoprotein metabolism as suspected by the hitherto well-established functions of low-density lipoprotein receptor, in a variety of tissues. Recent studies using receptor-deficient or -overexpressing animals and cells have suggested that certain LRs are important regulators of the migration (and proliferation) of vascular smooth muscle cells (SMCs). LR expression is markedly induced in intimal or medial SMCs during the formation of atherosclerotic lesions. Because LRs can modulate the activity of the urokinase-type plasminogen activator (uPA) receptor and possibly of the platelet-derived growth factor (PDGF) receptor, LRs may influence the migration of SMCs through functional modulation of these membrane receptors. Therefore, SMC migration may be regulated by time-restricted expression of LRs. In agreement with the concept of functional interaction between LRs and membrane signaling receptors, a negative regulator of uPA receptor protein catabolism, LR11, has been identified. Statins modulate the PDGF-induced migration of intimal SMCs via the LR11/uPA receptor cascade. Selective modification of the LRs/uPA receptor/PDGF receptor systems in SMCs may be important for suppression of atherosclerotic plaque formation as well as for preventing intimal thickening after angioplasty.

LDL receptor family members (LRs) regulate the catabolism of membrane-associated proteins and are expressed in SMCs of atherosclerostic lesions. LRs modulate the activity of the urokinase-type plasminogen activator (uPA) receptor and possibly of the PDGF receptor. Selective modification of the LRs/membrane receptor system may be important for suppression of atherosclerosis.

【关键词】  LDL receptor family smooth muscle cells migration LR urokinasetype plasminogen activator receptor PDGF receptor


Introduction


The members of the low-density lipoprotein receptor family (LRs) are characterized by distinct functional domains present in characteristic numbers and arrangements ( Figure 1 ). The common structural domains in most LRs are the so-called low-density lipoprotein (LDL) receptor ligand binding repeats (type A), epidermal growth factor precursor homology repeats (type B1 and B2), epidermal growth factor precursor homology repeats with a consensus tetrapeptide, Tyr-Trp-Thr-Asp, and in the cytoplasmic region, signals for receptor internalization via coated pits. These LRs discovered to date are the LDL receptor, LDL receptor-related protein-1 (LRP-1), megalin, the very low-density lipoprotein (VLDL) receptor/LR8, apolipoprotein E receptor 2/LR8B, LR11, and, most recently, LRPs 3 through 7. 1-3 LRP-1 and megalin are giant LRs in which the amino acid sequence contains multiple repeats of each functional component of the LDL receptor. 4,5 The domain structures of VLDL receptor/LR8 and apolipoprotein E receptor 2/LR8B are most similar to that of the LDL receptor. 6-8 LRs indeed show considerable sequence identity (70% to 100%) between molecules harboring common structures and among a wide range of species. Such sequence conservation is thought to indicate evolution from an ancestral gene by duplication or exon shuffling. The avian VLDL receptor/LR8 is essential for reproduction as a receptor for the yolk accumulation. 8,9


Figure 1. Schematic presentation of the LRs. The common structural modules in most of LRs are: (1) the so-called "LDL receptor ligand binding repeats (type A)," complement-type domains consisting of 40 residues displaying a triple-disulfide-bond-stabilized negatively charged surface; (2) epidermal growth factor (EGF) precursor homology repeats (type B1 and B2), also containing 6 cysteins each; (3) EGF precursor homology repeats consisting of 50 residues each, most often in groups of 5, with a consensus tetrapeptide, Tyr-Trp-Thr-Asp (YWTD); and (4) in the cytoplasmic region, signals for receptor internalization via coated pits, containing the consensus tetrapeptide Asn-Pro-Xaa-Tyr (NPXY).


LRs play a key role in lipoprotein metabolism, as demonstrated by the well-established actions of the LDL receptor in a variety of tissues. 1 Extensive functional analyses have also revealed that LRs play an important role in the catabolism of many membrane-associated proteins such as complexes between proteinases and their receptors. 1-3 Recent studies using receptor-deficient or -overexpressing animals and cells have suggested that certain LRs are also important as regulators of the migration (and proliferation) of various cells such as fibroblasts, neurons, and vascular smooth muscle cells (SMCs). 10-17


Histochemical studies have revealed that the expression of LRs, as well as scavenger receptors, is markedly induced during the development of atherosclerotic lesions. 1,18 For instance, the VLDL receptor/LR8 is highly expressed by SMCs, macrophages, and endothelial cells in rabbit atherosclerotic lesions, whereas the LDL receptor is not abundant in arterial walls. 18,19 LRP-1 expression is also induced in atheromatous plaques. 18-20 We identified strong LR11 expression inside plaques, particularly by intimal SMCs located at the interface between intima and media. 21,22 In addition, LRP-1B is expressed by SMCs of the medial layer and in thickened intimal regions. 23 Thus, changes in the expression of LRs by vascular cells, particularly SMCs, may play a role in the development of atherosclerosis.


The migration and proliferation of SMCs, as well as extracellular matrix (ECM) production and catabolism by these cells, are important events in the development of atherosclerosis and intimal thickening after coronary angioplasty. 24 When thickening of the intima occurs, SMCs migrate from the media into the intima. During migration, SMCs acquire or lose various functions to perform the above-mentioned activities in the intima. 25,26 However, the mechanisms that control the migration of intimal SMCs have not been clarified because of the complex intracellular machinery and the interactions of numerous internal or external factors and signaling pathways. There is conclusive evidence that migration of SMCs from the media into the intima contributes to the formation of stable plaque. 27,28 Here, we focus on the role of LRs in regulating membrane receptor functions related to the migration of SMCs associated with atherosclerosis.


Platelet-Derived Growth Factor-Mediated Migratory Activity of Intimal SMCs


There is a distinct difference in migratory activity between cultured SMCs isolated from the intimal and medial layers of atherosclerotic aortas. 29 Cultured intimal SMCs differ from medial SMCs in many ways, including their morphology, proliferative potential, and gene expression. 29-31 The phenotypic modifications of SMCs that migrate to the intimal layer seem to contribute to an enhanced synthetic capacity, representing a mechanism that influences plaque stability. In fact, cultured intimal SMCs exhibit a phenotype resembling that of fetal or dedifferentiated SMCs. 25,26 Among the many genes involved in the process of phenotypic modification that occurs in the intima, 32,33 the expression of myosin heavy chain isoforms, such as SM1, SM2, and SMemb/nonmuscle myosic heavy chain-B (NMHC-B), has been well characterized. 25,26,34


Many factors may contribute to altering the migratory potential of SMCs in the intima, including changes of contact with the ECM and exposure to growth factors. Cultured SMCs tend to mimic these changes because primary cultured cells rapidly lose their differentiation markers and develop a synthetic phenotype. Conversely, SMCs grown in 3D cultures, such as a honeycomb structure, are able to retain the contractile phenotype. 35 Thus, various cell culture models have provided information about factors that influence the migration of intimal SMCs. Among them, sensitivity to growth factors (including platelet-derived growth factor ) is known to be important for inducing SMC migration. 25 PDGF-BB-mediated intracellular signals induce migration, which is commonly observed using a migration assay system such as Boyden?s chamber. The influence of PDGF-BB on the migration of SMCs is mediated by a specific membrane receptor: PDGF ß-receptor. 36 During the process of migration of SMCs from the media into the intima, one of the strongly expressed genes is PDGF ß-receptor, 37 which contributes to the migratory capacity of intimal SMCs. 38,39 The PDGF ß-receptor is highly expressed even in the media of diabetic models, which show accelerated plaque formation. 40,41 PDGF-BB negatively regulates the transcription of multiple genes in SMCs and thus modulates differentiation. 42 Accordingly, the switch that induces PDGF ß-receptor gene expression seems to be closely related to increasing the migratory capacity of intimal SMCs.


Urokinase and Its Receptor System Are Activated During SMC Migration


In addition to chemoattractants, several proteases and their inhibitors are involved in the migration of SMCs through the process of matrix degradation. 24 Local protease activation is important for enhancing the mobility of migrating cells, particularly for SMCs to migrate through the ECM to target sites in plaque or thickened intima. Thus, matrix metalloproteinases (MMPs) are integral for SMC migration into the intima. 24 Conversion of pro-MMPs to active MMPs, as well as MMP-9 expression, is mediated by urokinase-type plasminogen activator (uPA)-generated plasmin. 43,44 The resulting matrix degradation releases growth factors such as fibroblast growth factor-2 and latent transforming growth factor-ß, and these chemoattractants further promote the migration of SMCs. Thus, urokinase appears to be necessary for migration of SMCs through the surrounding ECM.


Both tissue-type plasminogen activator and uPA cleave plasminogen to release plasmin. Expression of tissue-type plasminogen activator and uPA is increased in atherosclerotic plaque, 45-47 and a study using knockout mice has revealed a role of uPA in the development of intimal hyperplasia. 48 Accordingly, uPA is thought to play an important role in the target-oriented movement of SMCs because its activation can be localized via binding to its receptor (the uPA receptor) on the cell surface. The receptor-mediated potentiation of protease activity for plasminogen also causes an increase of plasmin activation around cell surface receptors. Subsequent production of plasmin leads to the degradation of ECM components and also has the potential to activate some MMPs. The essential role of this process in enhancing cell mobility has been intensively studied with regard to tumor invasion and neuronal migration. 49,50


Expression of uPA by medial SMCs increases rapidly and significantly after balloon catheter injury to a vessel, corresponding with the time course of SMC migration. 51 Virally mediated overexpression of uPA by the endothelial cells of the carotid arteries promotes lesion growth in cholesterol-fed rabbits. 52 After arterial injury, intimal thickening is significantly reduced in uPA-deficient mice. 48,53 Thus, uPA itself seems to promote intimal thickening after vascular injury. However, despite the ability of uPA to influence the migration of cultured SMCs, 54-56 intimal formation is unaffected in uPA receptor knockout mice. 57 The specific proteolytic activity of uPA plays a role in the processes of arterial repair after injury, although the details of the mechanism regulating association with its receptor have not been clarified in the setting of atherosclerosis.


In addition to the proteolytic cascade initiated by binding of uPA to its cell surface receptor, uPA possibly facilitates cell migration by inducing intracellular signaling pathways. 58 The uPA receptor is a glycosylphosphatidylinositol-anchored protein, and therefore signaling activity is mediated by its interaction with other membrane molecules. Binding of uPA to its receptor on the cell surface influences the migratory activity through the formation of a complex involving the uPA receptor, vitronectin, and integrin. 50,58 These interactions at the cell membrane stimulate intracellular signaling cascades, as well as uPA receptor-mediated activation of extracellular proteolysis. 50,58 uPA stimulates the migration of SMCs via its receptor signaling cascade containing the Janus kinase, Tyk2, and phosphatidylinositol 3-kinase. Active GTP-bound forms of small GTPases (RhoA and Rac1) are the downstream targets for Tyk2 and phosphatidylinositol 3-kinase activation. Phosphorylation of myosin light chain is one of the end points of the uPA receptor-mediated signaling pathways. Observations suggesting a possible role of uPA (independent of ECM degradation) in cell migration have been reported so the uPA receptor may also modulate migration/invasion in a protease-independent manner. These findings, together with the results obtained in uPA receptor knockout mice, 57 have led to the conception that the uPA receptor modulates SMC migration through cooperation between extracellular proteolysis and intracellular signaling. Proteolysis of the ECM accelerates migration and is coordinated with adhesive and structural changes that promote cell motility, with both processes leading the cells to their targets in the plaques.


LRs Are Novel Modulators of uPA Receptor Function During PDGF-Mediated Migration of SMCs


Functional modulation of the uPA receptor through the pathways with participation of LRs has been established. 59 LRs are known to play an integral role in the catabolism of lipoproteins and of complexes between proteinases and their receptors. 2,3 A large member of the family, LRP-1, is involved in the intake of uPA receptors and uPA/uPA receptor complexes by cells for subsequent degradation or recycling. 60 Extensive studies have revealed that other LRs, such as VLDL receptor/LR8 12 and LRP-1B, 61 also have the capacity to catabolize uPA/uPA receptor complexes.


LRP-1 is involved in the internalization of the uPA/uPA receptor complex, in which formation is induced by plasminogen activator inhibitor-1, and this process is dependent on LRP-1. 10,11,62,63 LRP-1 is a large molecule composed of 2 subunits. Two NP X Y motifs exist in the intracellular domain of LRP-1, and these motifs are not only important for endocytosis but also for intracellular signaling through molecules such as Shc. 64-66 Inhibition of uPA receptor internalization increases cell surface uPA receptor expression and enhances cell motility. 10,16,63,67


Deficiency of LRP-1 in SMCs causes atherosclerosis, which is mediated by the modulation of intracellular PDGF signaling. 17 This is attributable to the influence of LRP-1 on PDGF ß-receptor signaling or metabolism, possibly because of a molecular interaction at the cell surface. 17,68-70 LRP-1B is the giant family member that is most similar to LRP-1; it also binds to the PDGF ß-receptor and modulates receptor-mediated signaling in SMCs. 23 These findings suggest that SMC migration might be regulated by the time-restricted expression of LRs, which determines the outcome of PDGF ß-receptor- and uPA receptor-mediated signaling. In accordance with the concept of functional interaction between LRs and membrane signaling receptors, LR11 has been identified by us and others as a negative regulator of protein catabolism for uPA receptor. 71,72 Previous histochemical studies have revealed that LRs are markedly induced during the development of atherosclerotic lesions. 1,18 Altered expression of LRP-1 and the uPA receptor possibly reflects the vascular response to injury. Upregulation of LRP-1 mRNA has been detected in the aortas of rabbits fed a high-cholesterol diet. 1,18 Both LRP-1 mRNA and protein are expressed in normal and atherosclerotic human arteries. 19,20 Increased vascular expression of the uPA receptor is observed in cholesterol-fed rabbits and human atherosclerotic arteries. 73 Because LRs are able to modulate uPA receptor activity and possibly PDGF receptor activity, LRs are expected to regulate the migration of SMCs through the functional modulation of these membrane receptors ( Figure 2 ).


Figure 2. Proposed model for the regulation of SMC migration by LRs through the uPA/uPA receptor system. The uPA/uPA receptor system induces cell migration through both increased degradation of the ECM and receptor-mediated intracellular signaling that promotes motility. uPA receptor expression is regulated by LRs such as LRP-1, VLDL receptor/LR8, and LRP-1B. SMCs in plaques produce LR11, which is localized on the cell surface and also secreted by the cells. LR11 binds to and interacts with the uPA receptor on the cell surface or on neighboring cells. Formation of this complex inhibits internalization of the uPA receptor via other LRs (LRP-1, LRP-1B, etc.) and thereby prevents its degradation and relocation, resulting in the enhanced uPA receptor expression on the cell surface. Finally, SMCs expressing LR11 gain an increased migratory capacity that is mediated by activation of the uPA/uPA receptor system. LR11 gene transcription is induced by PDGF-BB and mediated by the PDGF ß-receptor. LRP-1 (and LRP-1B) interacts with the PDGF ß-receptor and modulates receptor-mediated intracellular signaling by PDGF-BB, which promotes migratory activity. Thus, LRs possibly regulate the migration of intimal SMCs in atherosclerotic plaques via modulation of PDGF receptor-meditated signaling, which is also linked with the uPA/uPA receptor system. Statins inhibit the migration of intimal SMCs by decreasing uPA receptor expression via the downregulation of LR11 gene expression.


Involvement of LRs in Regulating SMC Migration in the Intima


Recent functional studies using genetically altered animals or cells revealed that LRs are important regulators of the migration of various cells via modulation of cytokine signaling or protease activation. 13,16 SMC-specific inactivation of LRP-1 in mice has revealed a novel role of LRP-1, which forms a complex with the PDGF receptor. 17 LRP-1 ablation results in a decrease of vascular wall integrity and causes marked susceptibility to cholesterol-induced atherosclerosis in mice. 17 In murine embryonic fibroblasts and fibrosarcoma cells, loss of LRP-1 expression is associated with increased cell surface expression of the uPA receptor and is correlated with increased cell migration in vitro. 10 Similar changes were reported to occur when VLDL receptor/LR8 activity was neutralized in cultured breast cancer cells. 12 LR-mediated regulation of cell migration appears to depend partly on modulation of the uPA/uPA receptor system involved in the degradation of the ECM or modulation of uPA receptor-mediated intracellular signaling through activation of extracellular signal-regulated kinase and Rac1.


A negative regulator of receptor catabolism, LR11, controls uPA receptor localization on the plasma membrane because both the membrane-spanning and secreted forms of LR11 bind to and colocalize with the uPA receptor on the cell surface. 21,74 Expression of LR11 is induced by stimulation of PDGF-BB in SMCs and is observed in intimal SMCs localized at the intima/media border in the atherosclerotic plaques of experimental animals. 21 Overexpression of LR11 by SMCs enhances their migration by elevating uPA receptor expression. 21 Contrarily, neutralization of LR11 reduces the intimal thickening after cuff injury in mice. 21


Modulation of the LR11/uPAR Pathway for Prevention of Atherosclerosis


Statins are potent inhibitors of the 3-hydroxy-3-methylglutaryl-coenzyme A reductase that are known to be effective for preventing atherosclerosis. Statins have recently been shown to perform a multitude of activities that are involved in the functional modulation of vascular cells such as influences on cell proliferation and secretion. 75,76 One of the major effects of statins on SMCs is modulation of migration. However, the mechanism involved and clinical significance of such inhibition of migration, which has been observed in vitro, have not been elucidated. PDGF-induced migration of SMCs is suppressed by statins in vitro. 77,78 Statins reduce protease expression in atheromatous plaques, and hydrophilic statins decrease SMC numbers and collagen gene expression in vivo. 79 However, phenotypic modulation of intimal SMCs by statins has not yet been investigated. LR11 plays an important role in the induction of migration after enhancement by PDGF-BB in vitro. A potent 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor, pitavastatin, reduced the expression of both LR11 and SMemb/NMHC-B in atherosclerotic plaques (unpublished data, 2006). In fact, the enhanced expression of LR11, uPA receptor, and SMemb/NMHC-B by cultured intimal SMCs is reduced by pitavastatin to the levels seen in cells from the media. When expression of the uPA receptor, SMemb/NMHC-B, and endogenous LR11 is increased by PDGF-BB, the enhanced migratory activity of SMCs is blocked by pitavastatin via suppression of endogenous LR11 production. Thus, modulation of the LR11/uPA receptor system plays a role in PDGF-induced migration of intimal SMCs ( Figure 2 ).


It has not yet been clarified whether inhibition of the migration of intimal SMCs leads to the regression of atherosclerotic plaque or prevents restenosis after coronary angioplasty. Activation of pathways mediated by the uPA receptor and the PDGF receptor that increase the migration of intimal SMCs is thought to be essential for the formation of mature plaque after endothelial injury leads to the initiation of atherosclerosis. Unregulated expression of these membrane receptors may reduce the stability of plaque because the programmed migration of SMCs from the media to target regions in the intima would be disturbed. LRs are a possible candidate for modulating SMC migration to control the process of atherosclerosis. Selective modification of the LRs/uPA receptor/PDGF receptor system in SMCs, associated with the change to a dedifferentiated phenotype, appears to be important for the occurrence of intimal thickening after angioplasty as well as plaque formation in atherosclerosis.


Acknowledgments


This work was supported by grants from the Japanese Ministry of Education, Science, and Culture to Y.S. and H.B. We acknowledge that our research in this review was able to be performed because of help from numerous collaborators and colleagues. Finally, we thank Dr W.J. Schneider (University and Biocenter of Vienna) for his collaborative assistance over many years on this topic.

【参考文献】
  Schneider WJ, Nimpf J, Bujo H. Novel members of the LDL receptor superfamily and their potential roles in lipid metabolism. Curr Opin Lipidol. 1997; 8: 315-319.

Strickland DK, Gonias SL, Argraves WS. Diverse roles for the LDL receptor family. Trends Endocrinol Metab. 2002; 13: 66-74.

Herz J. The LDL receptor gene family: (un)expected signal transducers in the brain. Neuron. 2001; 29: 571-581.

Herz J, Hamann U, Rogne S, Myklebost O, Gausepohl H, Stanley KK. Surface location and high affinity for calcium of a 500-kd liver membrane protein closely related to the LDL-receptor suggest a physiological role as lipoprotein receptor. EMBO J. 1988; 7: 4119-4127.

Saito A, Pietromonaco S, Loo AK, Farquhar MG. Complete cloning and sequencing of rat gp330/"megalin," a distinctive member of the low density lipoprotein receptor gene family. Proc Natl Acad Sci U S A. 1994; 91: 9725-9729.

Takahashi S, Kawarabayasi Y, Nakai T, Sakai J, Yamamoto T. Rabbit very low density lipoprotein receptor: a low density lipoprotein receptor-like protein with distinct ligand specificity. Proc Natl Acad Sci U S A. 1992; 89: 9252-9256.

Kim DH, Iijima H, Goto K, Sakai J, Ishii H, Kim HJ, Suzuki H, Kondo H, Saeki S, Yamamoto T. Human apolipoprotein E receptor 2. A novel lipoprotein receptor of the low density lipoprotein receptor family predominantly expressed in brain. J Biol Chem. 1996; 271: 8373-8380.

Bujo H, Hermann M, Kaderli MO, Jacobsen L, Sugawara S, Nimpf J, Yamamoto T, Schneider WJ. Chicken oocyte growth is mediated by an eight ligand binding repeat member of the LDL receptor family. EMBO J. 1994; 13: 5165-5175.

Bujo H, Yamamoto T, Hayashi K, Hermann M, Nimpf J, Schneider WJ. Mutant oocytic low density lipoprotein receptor gene family member causes atherosclerosis and female sterility. Proc Natl Acad Sci U S A. 1995; 92: 9905-9909.

Weaver AM, Hussaini IM, Mazar A, Henkin J, Gonias SL. Embryonic fibroblasts that are genetically deficient in low density lipoprotein receptor-related protein demonstrate increased activity of the urokinase receptor system and accelerated migration on vitronectin. J Biol Chem. 1997; 272: 14372-14379.

Webb DJ, Nguyen DHD, Gonias SL. Extracellular signal-regulated kinase functions in the urokinase receptor-dependent pathway by which neutralization of low density lipoprotein receptor-related protein promotes fibrosarcoma cell migration and matrigel invasion. J Cell Sci. 2000; 113: 123-134.

Webb DJ, Nguyen DHD, Sankovic M, Gonias SL. The very low density lipoprotein receptor regulates urokinase receptor catabolism and breast cancer cell motility in vitro. J Biol Chem. 1999; 274: 7412-7420.

Trommsdorff M, Gotthardt M, Hiesberger T, Shelton J, Stockinger W, Nimpf J, Hammer RE, Richardson JA, Herz J. Reeler/Disabled-like disruption of neuronal migration in knockout mice lacking the VLDL receptor and ApoE receptor 2. Cell. 1999; 97: 689-701.

Zhu Y, Bujo H, Yamazaki H, Hirayama S, Kanaki T, Takahashi K, Shibasaki M, Schneider WJ, Saito Y. Enhanced expression of the LDL receptor family member LR11 increases migration of smooth muscle cells in vitro. Circulation. 2002; 105: 1830-1836.

Swertfeger DK, Bu G, Hui DY. Low density lipoprotein receptor-related protein mediates apolipoprotein E inhibition of smooth muscle cell migration. J Biol Chem. 2002; 277: 4141-4146.

Ma Z, Thomas KS, Webb DJ, Moravec R, Salicioni AM, Mars WM, Gonias SL. Regulation of Rac1 activation by the low density lipoprotein receptor-related protein. J Cell Biol. 2002; 159: 1061-1070.

Boucher P, Gotthardt M, Li WP, Anderson RG, Herz J. LRP: role in vascular wall integrity and protection from atherosclerosis. Science. 2003; 300: 329-332.

Ylä-Herttuala S. Expression of lipoprotein receptors and related molecules in atherosclerosis lesions. Curr Opin Lipidol. 1996; 7: 292-297.

Luoma JS, Hiltunen TP, Särkioja T, Moestrup SK, Gliemann J, Kodama T, Nikkari T, Ylä-Herttuala S. Expression of alpha2-macroglobulin receptor/low density lipoprotein receptor-related protein and scavenger receptor in human atherosclerotic lesions. J Clin Invest. 1994; 93: 2014-2021.

Hiltunen TP, Luoma JS, Nikkari T, Ylä-Herttuala S. Expression of LDL receptor, VLDL receptor, LDL receptor-related protein, and scavenger receptor in rabbit atherosclerotic lesions. Marked induction of scavenger receptor and VLDL receptor expression during lesion development. Circulation. 1998; 97: 1079-1086.

Zhu Y, Bujo H, Yamazaki H, Ohwaki K, Jiang M, Hirayama S, Kanaki T, Takahashi K, Shibasaki M, Schneider WJ, Saito Y. LR11, an LDL receptor gene family member, is a novel regulator of smooth muscle cell migration. Circ Res. 2004; 94: 752-758.

Kanaki T, Bujo H, Hirayama S, Ishii I, Morisaki N, Schneider WJ, Saito Y. Expression of LR11, a mosaic LDL receptor family member, is markedly increased in atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 1999; 19: 2687-2695.

Tanaga K, Bujo H, Zhu Y, Kanaki T, Hirayama S, Takahashi K, Inoue M, Mikami K, Schneider WJ, Saito Y. LRP1B attenuates the migration of smooth muscle cells by reducing membrane localization of urokinase and PDGF receptors. Arterioscler Thromb Vasc Biol. 2004; 24: 1422-1428.

Ross R. Atherosclerosis-an inflammatory disease. N Engl J Med. 1999; 340: 115-126.

Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev. 2004; 84: 767-801.

Sobue K, Hayashi K, Nishida W. Molecular mechanism of phenotypic modulation of smooth muscle cells. Horm Res. 1995; 50: 15-24.

Kumar MS, Owens GK. Combinatorial control of smooth muscle-specific gene expression. Arterioscler Thromb Vasc Biol. 2003; 23: 737-747.

Schwartz SM, deBlois D, O?Brien ER. The intima. Soil for atherosclerosis and restenosis. Circ Res. 1995; 77: 445-465.

Saito Y, Bujo H, Morisaki N, Shirai K, Yoshida S. Proliferation and LDL binding of cultured intimal smooth muscle cells from rabbits. Atherosclerosis. 1988; 69: 161-164.

Inaba T, Yamada N, Gotoda T, Shimano H, Shimada M, Momomura K, Kadowaki T, Motoyoshi K, Tsukada T, Morisaki N, Saito Y, Takaku F, Yazaki Y. Expression of M-CSF receptor encoded by c-fms on smooth muscle cells derived from arteriosclerotic lesion. J Biol Chem. 1992; 15;267: 5693-5699.

Koyama N, Harada K, Yamamoto A, Morisaki N, Saito Y, Yoshida S. Purification and characterization of an autocrine migration factor for vascular smooth muscle cells (SMC), SMC-derived migration factor. J Biol Chem. 1993; 268: 13301-13308.

Stolle K, Weitkamp B, Rauterberg J, Lorkowski S, Cullen P. Laser microdissection-based analysis of mRNA expression in human coronary arteries with intimal thickening. J Histochem Cytochem. 2004; 52: 1511-1518.

Mulvihill ER, Jaeger J, Sengupta R, Ruzzo WL, Reimer C, Lukito S, Schwartz SM. Atherosclerotic plaque smooth muscle cells have a distinct phenotype. Arterioscler Thromb Vasc Biol. 2004; 24: 1283-1289.

Manabe I, Nagai R. Regulation of smooth muscle phenotype. Curr Atheroscler Rep. 2003; 5: 214-222.

Ishii I, Tomizawa A, Kawachi H, Suzuki T, Kotani A, Koshushi I, Itoh H, Morisaki N, Bujo H, Saito Y, Ohmori S, Kitada M. Histological and functional analysis of vascular smooth muscle cells in a novel culture system with honeycomb-like structure. Atherosclerosis. 2001; 158: 377-384.

Ronnstrand L, Heldin CH. Mechanisms of platelet-derived growth factor-induced chemotaxis. Int J Cancer. 2001; 91: 757-762. <a href="/cgi/external_ref?access_num=10.1002/1097-0215(200002)9999:9999

Majesky MW, Reidy MA, Bowen-Pope DF, Hart CE, Wilcox JN, Schwartz SM. PDGF ligand and receptor gene expression during repair of arterial injury. J Cell Biol. 1990; 111: 2149-2158.

Seki N, Bujo H, Jiang M, Tanaga K, Takahashi K, Yagui K, Hashimoto N, Schneider WJ, Saito Y. LRP1B is a negative modulator of increased migration activity of intimal smooth muscle cells from rabbit aortic plaques. Biochem Biophys Res Commun. 2005; 331: 964-970.

Buetow BS, Tappan KA, Crosby JR, Seifert RA, Bowen-Pope DF. Chimera analysis supports a predominant role of PDGFRbeta in promoting smooth-muscle cell chemotaxis after arterial injury. Am J Pathol. 2003; 163: 979-984.

Inaba T, Ishibashi S, Gotoda T, Kawamura M, Morino N, Nojima Y, Kawakami M, Yazaki Y, Yamada N. Enhanced expression of platelet-derived growth factor-beta receptor by high glucose. Involvement of platelet-derived growth factor in diabetic angiopathy. Diabetes. 1996; 45: 507-512.

Tamura K, Kanzaki T, Tashiro J, Yokote K, Mori S, Ueda S, Saito Y, Morisaki N. Increased atherogenesis in Otsuka Long-Evans Tokushima fatty rats before the onset of diabetes mellitus: association with overexpression of PDGF beta-receptors in aortic smooth muscle cells. Atherosclerosis. 2000; 149: 351-358.

Dandre F, Owens GK. Platelet-derived growth factor-BB and Ets-1 transcription factor negatively regulate transcription of multiple smooth muscle cell differentiation marker genes. Am J Physiol Heart Circ Physiol. 2004; 286: H2042-H2051.

Lijnen HR. Plasmin and matrix metalloproteinases in vascular remodeling. Thromb Haemost. 2001; 86: 324-333.

Galis ZS, Khatri JJ. Matrix metalloproteinases in vascular remodeling and atherogenesis: the good, the bad, and the ugly. Circ Res. 2002; 90: 251-262.

Kienast J, Padro T, Steins M, Li CX, Schmid KW, Hammel D, Scheld HH, van de Loo JC. Relation of urokinase-type plasminogen activator expression to presence and severity of atherosclerotic lesions in human coronary arteries. Thromb Haemost. 1998; 79: 579-586.

Lupu F, Heim DA, Bachmann F, Hurni M, Kakkar VV, Kruithof EK. Plasminogen activator expression in human atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 1995; 15: 1444-1455.

Raghunath PN, Tomaszewski JE, Brady ST, Caron RJ, Okada SS, Barnathan ES. Plasminogen activator system in human coronary atherosclerosis. Arterioscler Thromb Vasc Biol. 1995; 15: 1432-1443.

Carmeliet P, Moons L, Herbert JM, Crawley J, Lupu F, Lijnen R, Collen D. Urokinase but not tissue plasminogen activator mediates arterial neointima formation in mice. Circ Res. 1997; 81: 829-839.

Kjoller L. The urokinase plasminogen activator receptor in the regulation of the actin cytoskeleton and cell motility. Biol Chem. 2002; 383: 5-19.

Ossowski L, Aguirre-Ghiso JA. Urokinase receptor and integrin partnership: coordination of signaling for cell adhesion, migration and growth. Curr Opin Cell Biol. 2000; 12: 613-620.

Clowes AW, Clowes MM, Au YP, Reidy MA, Belin D. Smooth muscle cells express urokinase during mitogenesis and tissue-type plasminogen activator during migration in injured rat carotid artery. Circ Res. 1990; 67: 61-67.

Falkenberg M, Tom C, DeYoung MB, Wen S, Linnemann R, Dichek DA. Increased expression of urokinase during atherosclerotic lesion development causes arterial constriction and lumen loss, and accelerates lesion growth. Proc Natl Acad Sci U S A. 2002; 99: 10665-10670.

Schafer K, Konstantinides S, Riedel C, Thinnes T, Muller K, Dellas C, Hasenfuss G, Loskutoff DJ. Different mechanisms of increased luminal stenosis after arterial injury in mice deficient for urokinase- or tissue-type plasminogen activator. Circulation. 2002; 106: 1847-1852.

Okada SS, Grobmyer SR, Barnathan ES. Contrasting effects of plasminogen activators, urokinase receptor, and LDL receptor-related protein on smooth muscle cell migration and invasion. Arterioscler Thromb Vasc Biol. 1996; 16: 1269-1276.

Herbert JM, Lamarche I, Carmeliet P. Urokinase and tissue-type plasminogen activator are required for the mitogenic and chemotactic effects of bovine fibroblast growth factor and platelet-derived growth factor-BB for vascular smooth muscle cells. J Biol Chem. 1997; 272: 23585-23591.

Wijnberg MJ, Quax PH, Nieuwenbroek NM, Verheijen JH. The migration of human smooth muscle cells in vitro is mediated by plasminogen activation and can be inhibited by alpha2-macroglobulin receptor associated protein. Thromb Haemost. 1997; 78: 880-886.

Carmeliet P, Moons L, Dewerchin M, Rosenberg S, Herbert JM, Lupu F, Collen D. Receptor-independent role of urokinase-type plasminogen activator in pericellular plasmin and matrix metalloproteinase proteolysis during vascular wound healing in mice. J Cell Biol. 1998; 140: 233-245.

Wei Y, Eble JA, Wang Z, Kreidberg JA, Chapman HA. Urokinase receptors promote beta1 integrin function through interactions with integrin alpha3beta1. Mol Biol Cell. 2001; 12: 2975-2986.

Herz J, Hui DY. Lipoprotein receptors in the vascular wall. Curr Opin Lipidol. 2004; 15: 175-181.

Nykjaer A, Conese M, Christensen EI, Olson D, Cremona O, Gliemann J, Blasi F. Recycling of the urokinase receptor upon internalization of the uPA:serpin complexes. EMBO J. 1997; 16: 2610-2620.

Li Y, Knisely JM, Lu W, McCormick LM, Wang J, Henkin J, Schwartz AL, Bu G. Low density lipoprotein (LDL) receptor-related protein 1B impairs urokinase receptor regeneration on the cell surface and inhibits cell migration. J Biol Chem. 2002; 277: 42366-42371.

Conese M, Nykjaer A, Petersen CM, Cremona O, Pardi R, Andreasen PA, Gliemann J, Christensen EI, Blasi F. Alpha-2 macroglobulin receptor/LDL receptor-related protein (LRP)-dependent internalization of the urokinase receptor. J Cell Biol. 1995; 13: 1609-1622.

Kjoller L, Hall A. Rac mediates cytoskeletal rearrangements and increased cell motility induced by urokinase-type plasminogen activator receptor binding to vitronectin. J Cell Biol. 2001; 152: 1145-1157.

Gotthardt M, Trommsdorff M, Nevitt MF, Shelton J, Richardson JA, Stockinger W, Nimpf J, Herz J. Interactions of the low density lipoprotein receptor gene family with cytosolic adaptor and scaffold proteins suggest diverse biological functions in cellular communication and signal transduction. J Biol Chem. 2000; 275: 25616-25624.

Barnes H, Larsen B, Tyers M, van Der Geer P. Tyrosine-phosphorylated low density lipoprotein receptor-related protein 1 (LRP-1) associates with the adaptor protein SHC in Src-transformed cells. J Biol Chem. 2001; 276: 19119-19125.

Su HP, Nakada-Tsukui K, Tosello-Trampont AC, Li Y, Bu G, Henson PM, Ravichandran KS. Interaction of CED-6/GULP, an adapter protein involved in engulfment of apoptotic cells with CED-1 and CD91/low density lipoprotein receptor-related protein (LRP). J Biol Chem. 2002; 277: 11772-11779.

Nguyen DH, Catling AD, Webb DJ, Sankovic M, Walker LA, Somlyo AV, Weber MJ, Gonias SL. Myosin light chain kinase functions downstream of Ras/ERK to promote migration of urokinase-type plasminogen activator-stimulated cells in an integrin-selective manner. J Cell Biol. 1999; 146: 149-164.

Boucher P, Gotthardt M. LRP and PDGF signaling: a pathway to atherosclerosis. Trends Cardiovasc Med. 2004; 14: 55-60.

Wu L, Arandjelovic S, Gonias SL. Effects of low density lipoprotein receptor-related protein-1 on the expression of platelet-derived growth factor beta-receptor in vitro. J Cell Biochem. 2004; 93: 1169-1177.

Takayama Y, May P, Anderson RG, Herz J. Low density lipoprotein receptor-related protein 1 (LRP1) controls endocytosis and c-CBL-mediated ubiquitination of the platelet-derived growth factor receptor {beta} (PDGFR{beta}). J Biol Chem. 2005; 280: 18504-18510.

Yamazaki H, Bujo H, Kusunoki J, Seimiya K, Kanaki T, Morisaki N, Schneider WJ, Saito Y. Elements of neural adhesion molecules and a yeast vacuolar protein sorting receptor are present in a novel mammalian low density lipoprotein receptor family member. J Biol Chem. 1996; 271: 24761-24768.

Jacobsen L, Madsen P, Moestrup SK, Lund AH, Tommerup N, Nykjaer A, Sottrup-Jensen L, Gliemann J, Petersen CM. Molecular characterization of a novel human hybrid-type receptor that binds the 2 -macroglobulin receptor associated protein (RAP). J Biol Chem. 1996; 271: 31379-31383.

Noda-Heiny H, Daugherty A, Sobel BE. Augmented urokinase receptor expression in atheroma. Arterioscler Thromb Vasc Biol. 1995; 15: 37-43.

Gliemann J, Hermey G, Nykjaer A, Petersen CM, Jacobsen C, Andreasen PA. The mosaic receptor sorLA/LR11 binds components of the plasminogen-activating system and platelet-derived growth factor-BB similarly to LRP1 (low-density lipoprotein receptor-related protein), but mediates slow internalization of bound ligand. Biochem J. 2004; 381: 203-212.

Takemoto M, Liao JK. Pleiotropic effects of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors. Arterioscler Thromb Vasc Biol. 2001; 21: 1712-1729.

Libby P, Aikawa M. Stabilization of atherosclerotic plaques: new mechanisms and clinical targets. Nat Med. 2002; 8: 1257-1262.

Kitahara M, Kanaki T, Toyoda K, Miyakoshi C, Tanaka S, Tamaki T, Saito Y. NK-104, a newly developed HMG-CoA reductase inhibitor, suppresses neointimal thickening by inhibiting smooth muscle cell growth and fibronectin production in balloon-injured rabbit carotid artery. Jpn J Pharmacol. 1998; 77: 117-128.

Kohno M, Shinomiya K, Abe S, Noma T, Kondo I, Oshita A, Takeuchi H, Takagi Y, Yukiiri K, Mizushige K. Inhibition of migration and proliferation of rat vascular smooth muscle cells by a new HMG-CoA reductase inhibitor, pitavastatin. Hypertens Res. 2002; 25: 279-285.

Fukumoto Y, Libby P, Rabkin E, Hill CC, Enomoto M, Hirouchi Y, Shiomi M, Aikawa M. Statins alter smooth muscle cell accumulation and collagen content in established atheroma of Watanabe heritable hyperlipidemic rabbits. Circulation. 2001; 103: 993-999.


作者单位:Departments of Genome Research and Clinical Application (H.B.), and Clinical Cell Biology (Y.S.), Chiba University Graduate School of Medicine, Japan.

作者: Hideaki Bujo; Yasushi Saito
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具