当前位置: 首页 > 期刊 > 《动脉硬化血栓血管生物学》 > 2003年第1期 > 正文
编号:10582265
Cardiac Ankyrin Repeat Protein(CARP) Expression in Human and Murine Atherosclerotic Lesions
http://www.100md.com 《动脉硬化血栓血管生物学》2003年第1期
     Abstract&5.(0c;, 百拇医药

    Objective— Cardiac ankyrin repeat protein (CARP) is a transcription factor-related protein that has been studied most extensively in the heart. In the present study, we investigated the expression and the potential function of CARP in human and murine atherosclerosis.&5.(0c;, 百拇医药

    Methods and Results— CARP expression was observed by in situ hybridization in endothelial cells lining human atherosclerotic plaques, whereas lesion macrophages were devoid of CARP. Furthermore, we established that CARP mRNA and smooth muscle (SM) actin antigen both colocalized in a subset of intimal smooth muscle cells (SMCs), whereas no CARP mRNA was encountered in quiescent SMCs in the media. The CARP mRNA-expressing intimal SMCs were distinct from intimal SMCs that synthesized the activation marker osteopontin or proliferating cell nuclear antigen. In addition, we showed that activin A, a member of the TGFß superfamily that prevents SMC-rich lesion formation, induced CARP mRNA expression in cultured SMCs.

    Conclusions— Based on our data and the knowledge that CARP reduces the proliferation of cultured SMCs, we propose that CARP is involved in inhibition of vascular lesion formation.;jx, 百拇医药

    Key Words: cardiac ankyrin repeat protein endothelial cells smooth muscle cells activin A atherogenesis;jx, 百拇医药

    Introduction;jx, 百拇医药

    Cardiac ankyrin repeat protein (CARP) was originally identified in vascular endothelial cells, cardiomyocytes, skeletal muscle and, more recently, in vascular smooth muscle cells (SMCs).1–8 The presence of nuclear localization signals and multiple ankyrin repeats indicates that CARP is a transcription factor-related protein that is involved in protein-protein interactions. Identification of the sequence of CARP mRNA and protein revealed mRNA decay elements in the 3'-untranslated region and a PEST-rich region in the protein.1–5 These structural elements are involved in rapid mRNA and protein degradation, which is a typical feature of early response genes.

    Until now, the function of CARP has been studied most extensively in the heart. Increased CARP expression is found in hypertrophy of the heart under physiological conditions (cardiogenesis) or pathophysiological conditions.3–7 Even though there are conflicting data on modulation of cardiac gene expression by CARP, a detailed analysis revealed that CARP prevents the activation of the MLC-2v promoter by binding the transcription factor YB-1 and consequently inhibits the activity of YB-1.4 Regulation of CARP expression in the heart is at least in part directed by the transcription factors Nkx2.5 and GATA-4, which are known to cooperate in the regulation of various cardiac genes.6,9–11 In skeletal muscle, CARP expression is induced in subcompartments of the muscle during normal morphogenesis or after denervation.2 In addition, in a myogenic cell line, CARP expression was upregulated on differentiation of these cells.2 In summary, CARP has been proposed to play a role in heart and skeletal muscle (re)differentiation.

    During atherosclerosis, endothelial cells and SMCs become activated and play a key role in intima formation. Activated endothelial cells attract inflammatory cells, and SMCs proliferate and migrate into the subendothelial space. A potential role for CARP in atherosclerosis is suggested from the knowledge that endothelial cells and SMCs synthesize CARP on stimulation. Specifically, in cultured endothelial cells, inflammatory cytokines, such as interleukin-1 and tumor necrosis factor-{alpha} , have been shown to increase CARP expression.1 In cultured SMCs, CARP expression is enhanced by transforming growth factor (TGF)-ß via the Smad signal transduction pathway, involving a TGF-ß-response element in the CARP promoter. Importantly, adenovirus-mediated overexpression of CARP results in decreased proliferation of SMCs.8 Furthermore, CARP mRNA synthesis increased in balloon-injured rat aorta, as determined by reverse-transcription polymerase chain reaction (RT-PCR). Collectively, these data suggest a role for CARP during atherogenesis.8 However, the cellular origin and exact localization of CARP expression in the (atherosclerotic) vessel wall has not been assessed. Therefore, we have investigated CARP expression in human and murine atherosclerotic lesions and demonstrate that only a subset of endothelial cells and SMCs express CARP. We have recently shown that activin A, a TGF-ß family member, prevents the formation of SMC-rich vascular lesions by maintaining the contractile SMC phenotype.12 In the current study we show induction of CARP mRNA expression in cultured SMCs on stimulation with activin A and we hypothesize a role for CARP downstream of activin A in preventing lesion formation.

    Methodsmoy)%ew, 百拇医药

    Human and Murine Tissue Samplesmoy)%ew, 百拇医药

    Human vascular tissue of atherosclerotic lesions was acquired during vascular surgery. Informed consent was obtained from patients according to protocols of the Medical Ethical Committee of the Academic Medical Center of Amsterdam. Tissue samples were fixed within 15 minutes after resection in 3.8% (v/v) formaldehyde in PBS and were subsequently paraffin-embedded.moy)%ew, 百拇医药

    Murine vascular tissues were obtained from APOE*3-Leiden mice that were fed a Western-type (ie, high-fat/cholesterol) diet, resulting in the formation of macrophage-derived foam cell-rich lesions that contained limited numbers of intimal SMCs, as has been described by van Vlijmen et al.13 After 16 to 24 weeks of this Western-type diet, the mice were sacrificed, and hearts and aortas were harvested. Sections were taken at the aortic valves. At this time point, blood cholesterol levels of the APOE*3-Leiden mice were 38.5±7.8 mmol/L, in contrast to 6.9±0.2 mmol/L in control mice. In addition, murine lesions were obtained from mice of a C57Bl6 genetic background that underwent carotid artery ligation. The carotid artery ligation model has been developed by Kumar and Lindner and results in the induction of SMC-rich lesions containing hardly any macrophages in the intima.14 Murine tissues were fixed by perfusion fixation with 4% (v/v) paraformaldehyde in PBS and were paraffin-embedded after resection. The sections (5 µm) were mounted on SuperFrost Plus glass slides.

    Immunohistochemistrywk{67, http://www.100md.com

    Antibody 1A4 (DAKO) recognizes human and murine SM {alpha} -actin and was used to detect SMCs, HAM56 (DAKO) recognizes human macrophages, and PC10 detects proliferating cell nuclear antigen (PCNA) (DAKO). Immunohistochemistry was performed on sections that were rehydrated and blocked with 10% (v/v) preimmune goat serum (DAKO). Subsequently, the sections were incubated overnight at 4°C with specific antibodies in Tris-buffered saline (10 mmol/L Tris-HCl, pH 8.0, 150 mmol/L NaCl), followed by incubations with biotinylated secondary antibodies that were detected with streptavidin-horseradish peroxidase conjugates (DAKO). Peroxidase activity was visualized with amino-ethyl carbazole and hydrogen peroxide. The PCNA protocol involved antigen retrieval in 10 mmol/L citrate buffer at pH 6.0. The sections were counterstained with hematoxylin, except for the PCNA staining.wk{67, http://www.100md.com

    In Situ Hybridizationwk{67, http://www.100md.com

    Riboprobes were synthesized and radiolabeled with [35S]-UTP (Amersham). For human CARP, a probe corresponding with the 454- to 1261-bp fragment of GenBank No. was used. For murine CARP, bp 218 to 872 of GenBank No. was used, and for murine osteopontin, bp 1 to 663 of GenBank No. was used as probes. Both antisense and sense riboprobes were assayed, of which the latter ones served as controls for specificity of the signal. In situ hybridization was performed by standard procedures.15

    To combine in situ hybridization and immunohistochemistry, human sections were blocked with 10% (v/v) preimmune goat serum in Tris-buffered saline after the last washing step of the in situ hybridization protocol. Thereafter, antibodies were applied, and the immunohistochemical protocol was continued. After visualization of peroxidase activity, the sections were covered with nuclear research emulsion (ILFORD Imaging UK Limited) and exposed. After the slides were developed, these double-stained sections were counterstained with hematoxylin. In all in situ hybridization experiments, for each specimen 3 to 6 consecutive sections were applied.-.t(*26, http://www.100md.com

    Cell Culture-.t(*26, http://www.100md.com

    SMCs were obtained from human saphenous vein or iliac artery explant cultures and maintained at 37°C in a humidified 5% CO2 chamber in 40% (v/v) Medium-199 with L-glutamine/L-amino acids, 40% (v/v) RPMI 1640 HEPES-buffered/L-glutamine, and 20% (v/v) human serum supplemented with penicillin and streptomycin (GIBCO BRL). Cultured SMCs were characterized by immunofluorescence with the anti-SM {alpha} -actin 1A4 antibody.15 The SMCs show uniform fibrillar staining. Confluent SMC cultures were made quiescent by incubation overnight in serum-free medium, supplemented with insulin (5 µg/mL), transferrin (5 µg/mL), selenite (5 ng/mL; GIBCO-BRL), and vitamin C (0.2 mmol/L). Subsequently, 100 ng/mL recombinant human activin A and 10 ng/mL TGFß (Roche Diagnostics) was added to the culture medium during 0, 2, 8, 24, and 48 hours of the venous SMCs. The arterial SMCs were stimulated with various concentrations activin A (0, 10, 50, and 100 ng/mL) for 6 hours. Purified, recombinant human activin A (lot #15365 to 361) was obtained from Dr. Pawson through the National Hormone and Pituitary Program, the National Institute of Diabetes and Digestive and Kidney Disease, and the US Department of Agriculture (Bethesda, Md).

    RNA Isolation and Northern Blotting-#k[, http://www.100md.com

    Total RNA was isolated from cultured cells by using the TRIzolTM method (GIBCO-BRL). Ten micrograms of total RNA per lane was subjected to electrophoresis on a formaldehyde-agarose gel and transferred to Hybond nylon membranes (Amersham). The blot was prehybridized at 42°C in formamide-containing buffers according to standard procedures. Probes were radiolabeled with [32P]-{alpha} ATP (Amersham) applying the random primer labeling mix (GIBCO BRL). To detect human CARP mRNA, a cDNA probe of 807 bp (454- to 1261-bp fragment of GenBank No. ) was applied and for GAPDH a fragment of 65 bp (480 to 545 bp of GenBank No. ). SM {alpha} -actin and SM22{alpha} mRNA were detected with a radiolabeled oligonucleotide and a random hexamer-radiolabeled cDNA probe, respectively, as has been described previously.15-#k[, http://www.100md.com

    RT-PCR-#k[, http://www.100md.com

    cDNA was generated by reverse transcription of RNA samples by using oligo dT primers and Superscript II (GIBCO-BRL). CARP and hypoxanthine guanine phosphoribosyltransferase (HPRT) sequences were amplified from the cDNA samples with Taq DNA polymerase (Amersham) and transcript specific primers sets (CARP: 5'-GGGCCAACTCCAGGGATTCC-3', rev: 5'-GGCCATGCCTTCAAAATGCCA-3' and HPRT; fwd: 5'-TAATTATGGACAGGACTGAACG-3', rev: 5'-CACAATCAAGACATTCTTTCCAG-3'). A standard PCR program with 65°C as annealing temperature resulted after 35 cycles in CARP and HPRT-specific products that were analyzed by agarose gel electrophoresis.

    Results3jv'a{%, http://www.100md.com

    CARP Expression in Human Atherosclerotic Lesions3jv'a{%, http://www.100md.com

    To substantiate a potential function of CARP in human atherogenesis, we determined the exact expression pattern of CARP mRNA by in situ hybridization in human atherosclerotic lesions derived from aorta or iliac artery. We assayed 16 specimens derived from individuals ranging in age from 38 to 65 years with lesions varying from type II to type V that were classified according to guidelines from the American Heart Association16 (). Of all lesions examined, only one lesion (type II, aorta, specimen #1 in ) did not show any CARP-positive endothelial cells or SMCs. CARP is expressed in up to 50% of the endothelial cells overlaying early type II lesions as well as in endothelial cells on advanced type IV-V lesions (see online Figure I, which can be accessed at http://atvb.ahajournals.org).3jv'a{%, http://www.100md.com

    fig.ommitted3jv'a{%, http://www.100md.com

    CARP Expression in Endothelial Cells and SMCs in Vascular Specimens

    fig.ommitted8+7:', 百拇医药

     CARP mRNA is expressed in human intimal SMCs but not in macrophages. Two serial sections of an advanced human lesion, derived from the iliac artery (see Table 1, #15), were analyzed simultaneously by immunohistochemistry and by in situ hybridization to identify CARP-expressing cells. In A through C, in situ hybridization to detect CARP mRNA was combined with SMC-specific immunohistochemistry, whereas in D through F, CARP mRNA expression and a macrophage-specific antigen were detected within the same section. CARP mRNA expression (black silver grains) colocalizes with some intimal SMCs (in red) as shown in the enlarged boxes 1 and 2 (B and C), whereas macrophages (D through F, in red) do not express CARP (higher magnifications of box 3 and 4 shown in E and F). Original magnification x50 (A and D) and x630 (B, C, E, and F). M indicates macrophages; Ad, adventitia; Int, intima; Lu, lumen; and M, media.8+7:', 百拇医药

    To assess the expression profile of CARP in SMCs and macrophages, two consecutive sections were assayed for CARP mRNA and simultaneously stained with specific antibodies to detect either SMCs (C) or macrophages (F). Obviously, the CARP-positive cells, lining the lumen of the vessel, are CARP-expressing endothelial cells (F). The intima of this advanced lesion contains a thin fibrous cap, a large macrophage infiltrate, extracellular lipid pools, and a small acellular core. Colocalization of CARP mRNA (black dots) and the SMC marker SM {alpha} -actin (red) show that CARP is expressed in a subset of intimal SMCs (C). CARP-positive intimal SMCs were seen in the fibrous cap (C) and in the SMC-rich shoulder of the lesion (see online Figure IIA, which can be accessed at http://atvb.ahajournals.org). CARP expression is absent in medial SMCs and, in addition, has never been observed in macrophages (illustrated inF).

    To establish whether CARP expression correlates with the proliferative state of SMCs in the intima, PCNA staining was performed (Figure II). The subset of CARP-expressing SMCs did not colocalize with the population of PCNA-positive cells in the intima, suggesting that CARP is not involved in SMC proliferation.i$z, http://www.100md.com

    CARP Expression in Murine Atherosclerotic and Restenotic Lesionsi$z, http://www.100md.com

    To further delineate the expression profile of CARP in atherogenesis, we analyzed CARP expression in murine diet-induced atherosclerotic lesions and in lesions formed in a restenosis model. In APOE*3-Leiden transgenic mice, a Western-type diet results in the formation of lesions containing lipid-laden macrophages. Interestingly, the occasional SMCs present in these lesions, which form a fibrous cap, do express CARP . Clearly, CARP mRNA colocalizes with SM {alpha} -actin as shown in a consecutive section . CARP is only expressed in intimal SMCs and was never observed in medial SMCs or macrophages in this murine model, which is similar to our observations with human atherosclerotic specimens. Determination of the number of CARP-positive SMCs at different time periods of Western-type diet revealed that CARP reaches optimal expression after 16 weeks of diet, which correlates with the increased presence of intimal SMCs in these lesions (see online Figure IIIA, which can be accessed at http://atvb.ahajournals.org).

    fig.ommitted-&'fw{&, 百拇医药

    . CARP mRNA expression in murine lesions. CARP mRNA expression was analyzed in an atherosclerotic lesion at the aortic valves of an APOE*3-Leiden mouse after 16 weeks of Western-type diet (A, arrows; box enlarged in B). SM -actin immunostaining (C) indicates that CARP is expressed in the few SMCs present in the macrophage-rich lesion in APOE*3-Leiden mice. CARP expression is shown in an SMC-rich lesion that was induced by carotid artery ligation (4 weeks; D). Osteopontin mRNA expression (E) and PCNA protein expression (F, gray arrows) do not colocalize with CARP expression (D). Dashed red lines highlight the internal elastic lamina between the intima (Int) and media (M) and the dotted red lines mark the lumen. Original magnification x100 (A), x200 (D through F), and x630 (B and C). Lu, lumen.-&'fw{&, 百拇医药

    To evaluate the expression of CARP in intimal SMCs in further detail, we studied its expression pattern in lesions generated in the murine carotid artery ligation model. In the SMC-rich lesions induced in this model, CARP mRNA expression is present in a subset of intimal SMCs, indicating that intimal SMCs do not form a homogenous population. To correlate CARP expression with a SMC phenotype, we analyzed in consecutive sections the expression of osteopontin and PCNA, which are markers for proliferative SMCs.17,18 In murine lesions, osteopontin may be considered a relatively broad activation marker, whereas PCNA is only expressed during a short period of the cell cycle. A typical lesion is presented , showing that the expression of CARP (D) does neither colocalize with osteopontin (E) nor with PCNA (F), which may indicate that CARP expression is not associated with intimal SMC proliferation. Analysis of temporal and spatial expression of CARP revealed that after 1 week of ligation, most CARP-positive cells are present in the media (Figure IIIB). At this time point, hardly any intima has been formed. Yet, medial SMCs are activated, express osteopontin (data not shown), and have started to proliferate and migrate to initiate the formation of a lesion. After 2.5 and 4 weeks of ligation, an extensive intima is present containing a subpopulation of CARP-positive SMCs. Already at 2.5 weeks, the medial SMCs are quiescent again and accordingly show only an occasional CARP-expressing SMC.

    Note that we do not observe CARP expressing endothelial cells in the murine lesions. This could be either the result of the low number of endothelial cells present in the murine lesions, a different timeframe of CARP expression in the mouse, or a species difference between mice and humans.|\, 百拇医药

    Effect of Activin A Stimulation on CARP Expression|\, 百拇医药

    CARP expression is induced in SMCs by TGF-ß via the Smad signal transduction pathway, which is mediated by a TGF-ß response element in the CARP promoter.8 Activin A is a member of the TGF-ß superfamily, and its signaling is mediated through the same Smad proteins as TGF-ß signaling. To assess a potential effect of activin A on CARP expression, we added purified activin A for 2 to 48 hours to cultured SMCs and performed Northern blotting analyses. The intensity of the bands, as determined by phosphorimager analysis and after correction for the GAPDH control, was plotted in line graphs (See online Figure IV, which can be accessed at http://atvb.ahajournals.org). Activin A increased steady-state CARP mRNA levels in SMCs, showing a 2.8-fold augmented expression from 8 to 48 hours. SM -actin and SM22 were continuously increasing (ultimately 10.8-fold and 2.9-fold, respectively) after 48 hours of activin A treatment, indicating the transition of the cellular phenotype into a contractile, nonproliferative SMC phenotype.15 TGF-ß was added to the cultured SMCs as a control, leading to similar increase in CARP and SM {alpha} -actin expression levels (both 2.9-fold). In addition, activin A stimulation of arterial SMCs induced CARP expression in a concentration-dependent manner as determined by RT-PCR . Induction of CARP expression was already observed at an activin A concentration of 10 ng/mL and was optimal at 50 ng/mL.

    fig.ommitted7'w&&^-, http://www.100md.com

     Activin A induces CARP expression in cultured SMCs. Cultured human SMCs were treated for 0, 2, 8, 24, and 48 hours with activin A or TGF-ß (8 hours). Total RNA was assayed by Northern blotting for CARP, SM -actin, SM22, and GAPDH mRNA expression (A). CARP expression, as determined by RT-PCR, was also increased on activin A stimulation (6 hours) in a dose-dependent way (0, 10, 50, and 100 ng/mL) (B). Optimal CARP induction was reached at a concentration of 50 ng/mL of activin A. HPRT was assayed as a control for equal cDNA input.7'w&&^-, http://www.100md.com

    Discussion7'w&&^-, http://www.100md.com

    In this study, we observed CARP expression in human and murine atherosclerotic lesions in endothelial cells and SMCs. Based on its mere presence in atherosclerotic lesions, CARP may be considered as a factor that is actively involved in this pathological process. Several arguments may be put forward to support the possibility that CARP restrains the formation of vascular lesions. First, CARP contains 4 ankyrin repeats, which are protein modules that are well studied in the I{kappa} B family.19 Ankyrin repeats in IB are functionally involved in binding and inhibition of the transcription factor nuclear factor B. In line with the inhibitory function of IB and in analogy to CARP binding of transcription factor YB-1 in cardiomyocytes,4 CARP may also be a negative regulator of transcription factors in endothelial cells and SMCs. Second, in human and murine lesions we have shown that CARP-expressing SMCs are a different subset of intimal SMCs than the subset of SMCs that express PCNA or osteopontin. Because PCNA and osteopontin have been associated with activated SMCs that are proliferative and migratory,17,18 CARP expression seems to be associated with quiescent intimal SMCs. It should be noted that CARP is not present in healthy vessels, and for that reason this protein is not simply a marker for quiescent SMCs. This is further illustrated by the presence of CARP in the media of murine lesions after 1 week of carotid artery ligation and the absence of expression after 2.5 and 4 weeks, at which time the medial SMCs have become quiescent again. Consequently, CARP may be involved in the transition of activated SMCs into a resting phenotype. Third, TGF-ß8 and activin A (this study) are potent inducers of CARP expression in SMCs. Particularly, activin A is known as an inhibitor of atherogenesis, which is demonstrated by prevention of lesion formation both in saphenous vein organ cultures and in the murine, loose-fitting cuff model.12 Activin A has been shown to promote the contractile phenotype of SMCs, as is illustrated by the induction of expression of SM-specific markers, such as SM -actin and SM22 on stimulation of cultured SMCs .12,15 Fourth, overexpression of CARP in SMCs resulted in the inhibition of DNA synthesis.8 Kanai et al8 have shown that CARP-mediated inhibitory effects on SMC proliferation correlate well with increased levels of cyclin-dependent kinase inhibitor p21 and reduction of phosphorylation of the retinoblastoma protein Rb, involved in growth arrest.

    Based on these data, we propose that the transient expression of CARP in intimal SMCs may be involved in preventing vascular lesion formation.4kj@, 百拇医药

    Received August 28, 2002; accepted September 6, 2002.4kj@, 百拇医药

    References4kj@, 百拇医药

    Chu W, Burns DK, Swerlick RA, Presky DH. Identification and characterization of a novel cytokine-inducible nuclear protein from human endothelial cells. J Biol Chem. 1995; 270: 10236–10245.4kj@, 百拇医药

    Baumeister A, Arber S, Caroni P. Accumulation of muscle ankyrin repeat protein transcript reveals local activation of primary myotube endcompartments during muscle morphogenesis. J Cell Biol. 1997; 139: 1231–1242.4kj@, 百拇医药

    Jeyaseelan R, Poizat C, Baker RK, Abdishoo S, Isterabad LB, Lyons GE, Kedes L. A novel cardiac-restricted target for doxorubicin; CARP, a nuclear modulator of gene expression in cardiac progenitor cells and cardiomyocytes. J Biol Chem. 1997; 272: 22800–22808.4kj@, 百拇医药

    Zou Y, Evans S, Chen J, Kuo H-C, Harvey RP, Chien KR. CARP, a cardiac ankyrin repeat protein, is downstream in the Nkx2-5 homeobox gene pathway. Development. 1997; 124: 793–804.

    Aihara Y, Kurabayashi M, Arai M, Kedes L, Nagai R. Molecular cloning of rabbit CARP cDNA and its regulated expression in adriamycin-cardiomyopathy. Biochim Biophys Acta. 1999; 1447: 318–324.3ld.x, 百拇医药

    Kuo H-C, Chen J, Ruiz-Lozano P, Zou Y, Nemer M, Chien KR. Control of segmental expression of the cardiac-restricted ankyrin repeat protein gene by distinct regulatory pathways in murine cardiogenesis. Development. 1999; 126: 4223–4234.3ld.x, 百拇医药

    Manning BS, Shotwell K, Mao L, Rockman HA, Koch WJ. Physiological induction of a ß-adrenergic receptor kinase inhibitor transgene preserves ß-adrenergic responsiveness in pressure-overload cardiac hypertrophy. Circulation. 2000; 102: 2751–2757.3ld.x, 百拇医药

    Kanai H, Tanaka T, Aihara Y, Takeda S-I, Kawabata M, Miyazono K, Nagai R, Kurabayashi M. Transforming growth factor-ß/Smad signaling induces transcription of the cell type-restricted ankyrin repeat protein CARP gene through CAGA motif in vascular smooth muscle cells. Circ Res. 2001; 88: 30–36.3ld.x, 百拇医药

    Durocher D, Charron F, Warren R, Schwartz RJ, Nemer M. The cardiac transcription factors Nkx2.5 and GATA-4 are mutual cofactors. EMBO J. 1997; 16: 5687–5696.

    Lee Y, Shioi T, Kasahara H, Jobe SM, Wiese R, Markham BE, Izumo S. The cardiac tissue-restricted homeobox protein Csx/Nkx2.5 physically associates with the zinc finger protein GATA-4 and cooperatively activates atrial natriuretic factor gene expression. J Mol Cell Biol. 1998; 18: 3120–3129.#.\u, 百拇医药

    Sepulveda JL, Belaguli N, Nigam V, Chen CY, Nemer M, Schwartz RJ. GATA-4 and Nkx-2.5 coactivate Nkx-2 DNA binding targets: role for regulating early cardiac gene expression. J Mol Cell Biol. 1998; 18: 3405–3415.#.\u, 百拇医药

    Engelse MA, Lardenoye JH, Neele JM, Grimbergen JM, De Vries MR, Lamfers ML, Pannekoek H, Quax PH, De Vries CJ. Adenoviral activin A expression prevents intimal hyperplasia in human and murine blood vessels by maintaining the contractile smooth muscle cell phenotype. Circ Res. 2002; 90: 1128–1134.#.\u, 百拇医药

    van Vlijmen BJ, van den Maagdenberg AM, Gijbels MJ, van der Boom H, HogenEsch H, Frants RR, Hofker MH, Havekes LM. Diet-induced hyperlipoproteinemia and atherosclerosis in apolipoprotein E3-Leiden transgenic mice. J Clin Invest. 1994; 93: 1403–1410.#.\u, 百拇医药

    Kumar A, Lindner V. Remodeling with neointima formation in the mouse carotid artery after cessation of blood flow. Arterioscler Thromb Vasc Biol. 1997; 17: 2238–2244.

    Engelse MA, Neele JM, van Achterberg TA, van Aken BE, van Schaik RH, Pannekoek H, de Vries CJ. Human activin-A is expressed in the atherosclerotic lesion and promotes the contractile phenotype of smooth muscle cells. Circ Res. 1999; 85: 931–939.-{, http://www.100md.com

    Stary HC, Chandler AB, Dinsmore RE, Fuster V, Glagov S, Insull W Jr, Rosenfeld ME, Schwartz CJ, Wagner WD, Wissler RW. A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis: a report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Arterioscler Thromb Vasc Biol. 1995; 15: 1512–1531.-{, http://www.100md.com

    Isoda K, Nishikawa K, Kamezawa Y, Yoshida M, Kusuhara M, Moroi M, Tada N, Ohsuzu F. Osteopontin plays an important role in the development of medial thickening and neointimal formation. Circ Res. 2002; 91: 77–82.-{, http://www.100md.com

    Bravo R. Synthesis of PCNA and its relation with DNA replication. Exp Cell Res. 1986; 163: 287–293.-{, http://www.100md.com

    Simeonidis S, Stauber D, Chen G, Hendrickson WA, Thanos D. Mechanisms by which IkappaB proteins control NF-kappaB activity. Proc Natl Acad Sci U S A. 1999; 96: 49–54.(Vivian de Waard Tanja A.E. van Achterberg Nicholas J. Beauchamp Hans Pannekoek Carlie J.M. de Vries)