• 沒有找到結果。

Molecular mechanism of progesterone-induced antiproliferation in rat aortic smooth muscle cells.

N/A
N/A
Protected

Academic year: 2021

Share "Molecular mechanism of progesterone-induced antiproliferation in rat aortic smooth muscle cells."

Copied!
7
0
0

加載中.... (立即查看全文)

全文

(1)

Endocrinology 2003 144:2785-2790 originally published online Apr 10, 2003; , doi: 10.1210/en.2003-0045

Wen-Sen Lee, Chao-Wei Liu, Shu-Hui Juan, Yu-Chih Liang, Pei-Yin Ho and Yi-Hsuan Lee

Smooth Muscle Cells

Molecular Mechanism of Progesterone-Induced Antiproliferation in Rat Aortic

Society please go to: http://endo.endojournals.org//subscriptions/

or any of the other journals published by The Endocrine Endocrinology

To subscribe to

(2)

Molecular Mechanism of Progesterone-Induced

Antiproliferation in Rat Aortic Smooth Muscle Cells

WEN-SEN LEE, CHAO-WEI LIU, SHU-HUI JUAN, YU-CHIH LIANG, PEI-YIN HO, AND YI-HSUAN LEE

Graduate Institutes of Medical Sciences (W.-S.L., C.-W.L.) and Cell and Molecular Biology (P.-Y.H.), and Departments of Physiology (W.-S.L., S.-H.J., Y.-H.L.) and Internal Medicine (Y.-C.L.), School of Medicine, Taipei Medical University, Taipei 110, Taiwan

Previously we demonstrated that progesterone at physiologic levels dose dependently inhibited cell proliferation in cul-tured rat aortic smooth muscle cells (RASMCs). However, the molecular mechanism underlying of progesterone-induced antiproliferation was not clear. Here we demonstrated that progesterone induced a reduction of the [3H]thymidine

incor-poration into RASMCs during the S-phase of the cell cycle. Western blotting analysis revealed that the protein levels of cyclin A, cyclin E, and cyclin-dependent-kinase (CDK) 2 but not cyclin D1 and CDK4 decreased after progesterone treat-ment, but those of CDK-inhibitory proteins, p21 and p27, in-creased. Immunoprecipitation showed that the formations of the CDK2-p21 and CDK2-p27 complex were increased and the

assayable CDK2 kinase activity was decreased in the proges-terone-treated RASMCs. In contrast, the formations of the CDK4-p21 and CDK4-p27 complex and the assayable CDK4 kinase activity were not changed significantly by progester-one treatment. Pretreatment of RASMCs with a p21 or p27 antisense oligonucleotide reduced the progesterone-induced inhibition of [3H]thymidine incorporation into RASMCs. In

conclusion, these data suggest that progesterone inhibits RASMCs proliferation by increasing the levels of p21 and p27 protein, which in turn inhibit CDK2 kinase activity, and fi-nally interrupt the cell cycle. (Endocrinology 144: 2785–2790, 2003)

A

THEROSCLEROSIS AND ITS complications, such as coronary artery disease, stroke, and peripheral vas-cular disease, remain the principal causes of death in devel-oped countries. Although the pathogenesis of atherosclerosis is not fully elucidated, one theory holds that atherosclerosis is a response of the vascular wall to injury (1– 4). In response to injury and various stimuli, the activated vascular endo-thelium produces cytokines and growth factors to promote the growth and migration of vascular smooth muscle cells, key events in the formation of atherosclerotic lesions in hu-mans. Thus, one of the most important goals in the study of prevention of atherosclerosis is to identify factors that inhibit vascular smooth muscle cell proliferation and migration.

In humans, the epidemiological studies showed that pre-menopausal women have a much lower mortality from ath-erosclerotic cardiovascular disease than men, suggesting that sex hormones might have a cardioprotective effect (5). This hypothesis was supported by the evidence that estrogen replacement reduces the incidence of cardiovascular diseases in postmenopausal women (6). In experimental animals, es-trogen administration inhibits the development of experi-mentally induced atherosclerosis (7–10). There has been little information on the effects of progesterone on cardiovascular disease. Previously, Grodstein and Stamper (11) showed that the relative risk of major coronary heart disease among post-menopausal women who took estrogen with progesterone was 0.39, compared with the risk of those who took estrogen alone, which was 0.60 (in which the risk among women who

took no hormones was set at 1.0). In castrated baboon re-ceiving estradiol and progesterone together had fewer vas-cular lesions than those receiving estradiol alone.

The exact mechanisms of the cardioprotective or athero-protective effect of sex hormones are not well understood. Smooth muscle cell proliferation and migration play an im-portant role in the genesis of the atherosclerotic plaque. Vas-cular smooth muscle cells normally reside in the media of the artery, have a low proliferative index, and are surrounded by a meshwork of several extracellular matrix components. However, in the process of atherogenesis, smooth muscle cell proliferation is increased in the forming neointima and in-nermost part of the underlying media (12). Previous studies have demonstrated that estrogen exerts a direct effect to cause vasodilation, suppress collagen synthesis, and act as calcium antagonist properties. Estrogen also exerts an indi-rect effect to inhibit low-density lipoprotein oxidation, at-tenuate coronary and aortic low-density lipoprotein accu-mulation, and increase plasma high-density lipoprotein levels (13). However, there is little evidence of a direct effect of sex hormones on proliferation of vascular smooth muscle cells. The localization of estrogen and progesterone receptors in the medial layer of aorta led us to hypothesize that sex hormones might have a direct effect on the proliferation of the vascular smooth muscle cell.

Previously we demonstrated that progesterone (the nat-ural hormone), but not estrogen, at physiologic levels inhib-ited DNA synthesis and decreased cell number in cultured rat and human aortic smooth muscle cells in a dose-depen-dent manner (14). However, the underlying mechanism of progesterone-induced antiproliferation was not clear. The findings of this study will provide important insights into the molecular and cellular mechanisms of atheroprotective

ef-Abbreviations: AS, Antisense oligonucleotide; CDK, cyclin-depen-dent kinase; CKI, CDK inhibitor; DTT, dithiothreitol; FACS, fluores-cence-activated cell sorter; FBS, fetal bovine serum; G3PDH, glycerol-3-phosphate dehydrogenase; RASMC, rat aortic smooth muscle cell.

doi: 10.1210/en.2003-0045

(3)

fects of progesterone. Only when the mechanism of athero-protective effects of progesterone is fully understood can we begin to design a strategy for preventing and treating ath-erosclerosis and its complications.

Materials and Methods

Materials

Water-soluble progesterone was purchased from Sigma (St. Louis, MO). Dithiothreitol (DTT), HEPES, EDTA, glycerol, phenylmethyl-sulfonyl fluoride, pepstatin A, leupeptin, sodium dodecyl sulfate (SDS), Nonidet P-40, trypsin-EDTA, and kanamycin were purchased from Life Technologies, Inc. (Carlsbad, CA). Fetal bovine serum (FBS) was purchased from Hyclone Laboratories, Inc. (Logan, UT). Anti-bodies specific for cyclins, cyclin-dependent-kinases (CDKs), and CDK inhibitors (CKIs) were purchased from Transduction Labora-tories, Inc. (Lexington, KY). An antibody specific for glycerol-3-phos-phate dehydrogenase (G3PDH) was purchased from Biogenesis (Kingston, NH). Antimouse IgG-conjugated alkaline phosphatase was purchased from Jackson ImmunoResearch Laboratories, Inc. (West Grove, PA). 4-Nitro blue tetrazolium and 5-bromo-4-chloro-3-indolyl-phosphate were purchased from Kirkegaard & Perry Lab-oratories (Gaithersburg, MD). Protein assay agents were purchased from Bio-Rad Laboratories, Inc. (Hercules, CA).

Cell culture

Rat aortic smooth muscle cells (RASMCs) were harvested from the thoracic aortas of adult male Sprague Dawley rats (200 –250 g) by en-zymatic dissociation. The cells were grown in DMEM supplemented with 10% FBS and penicillin (100 U ml⫺1), streptomycin (100␮g/ml), and 25 mm HEPES (pH 7.4) in a humidified 37 C incubator. After the cells had grown to confluence, they were disaggregated in trypsin solution, washed with DMEM containing 10% FBS, centrifuged at 125⫻ g for 5 min, resuspended, and then subcultured according to standard proto-cols. Cells from passages 5–9 were used.

[3H]Thymidine incorporation

As previously described (15, 16), RASMCs at a density of 1⫻ 104

cells/cm3were applied to 24-well plates in growth medium (DMEM

plus 10% FBS). After the cells had grown to 70 – 80% confluence, they were rendered quiescent by incubation for 72 h in DMEM containing 0.04% charcoal/dextran-treated FBS (Hyclone Laboratories, Inc.). Phe-nol red-free DMEM was used in the experiments with progesterone. Water-soluble progesterone or PBS in 2% FBS was added to the cells at various concentrations, and the mixture was allowed to incubate for 24 h. During the last 2 h of the incubation with or without progesterone, [3H]thymidine was added at 1␮Ci/ml⫺1(1␮Ci ⫽ 37 kBq). Incorporated

[3H]thymidine was extracted in 0.2 n NaOH and measured in a liquid

scintillation counter.

Protein preparation and Western blotting

To determine the expression levels of cyclins, CDKs, CKIs, and G3PDH in RASMCs, the total proteins were extracted, and Western blot analyses were performed as described previously (16). Briefly, RASMCs were cultured in 15-cm Petri dishes. After reaching subconfluence, the cells were rendered quiescent and then treated with various concentra-tions of progesterone for 24 h and incubated in a humidified incubator at 37 C. After incubation, the cells were washed with PBS (pH 7.4), incubated with extraction buffer (10 mm Tris, pH 7.0; 140 mm NaCl; 2 mm phenylmethylsulphonyl fluoride; 5 mm DTT; 0.5% Nonidet P-40; 0.05 mm pepstatin A; and 0.2 mm leupeptin) with gentle shaking and then centrifuged at 12,500⫻ g for 30 min. The cell extract was then boiled in a ratio of 1:1 with sample buffer (100 mm Tris, pH 6.8; 20% glycerol; 4% SDS; and 0.2% bromophenol blue). Electrophoresis was performed using 10% SDS-polyacrylamide gel (2 h, 110 V, 40 mA, 50␮g protein per lane). Separated proteins were transferred to polyvinyl difluoride mem-branes (3 h, 40 V), treated with 5% fat-free milk powder to block the nonspecific IgGs, and incubated for 1 h with specific antibody for cyclins, CDKs, CKIs, or G3PDH. The blot was then incubated with antimouse or

antirabbit IgG linked to alkaline phosphatase (1:1000) for 1 h. Subse-quently the membrane was developed with 4-nitro blue tetrazolium/ 5-bromo-4-chloro-3-indolyl-phosphate as a substrate.

Immunoprecipitation

As previously described (16), CDK2 or CDK4 was immunoprecipi-tated from 200␮g protein by using anti-CDK2 or anti-CDK4 antibody (2␮g) and protein A agarose beads (20 ␮l). The precipitates were washed five times with lysis buffer and once with PBS. The pellet was then resuspended in sample buffer (50 mm Tris, pH 6.8; 100 mm bromophenol blue; and 10% glycerol) and incubated at 90 C for 10 min before elec-trophoresis to release the proteins from the beads.

CDK assay

As previously described (17), CDK2 or CDK4 immunoprecipitates from progesterone-treated and control RASMCs were washed three times with lysis buffer and twice with kinase assay buffer (50 mm Tris-HCl, pH 7.4; 10 mm MgCl2; and 1 mm DTT). Phosphorylation of

histone H1 (for CDK2) and glutathione-S-transferase fusion protein (for CDK4) were measured by incubating the beads with 40␮l hot kinase solution [0.25␮l (2.5 ␮g) histone H1, 0.5 ␮l (␥-32P) ATP, 0.5␮l 0.1 mm

ATP, and 38.75␮l kinase buffer] at 37 C for 30 min. The reaction was stopped by boiling the sample in SDS sample buffer for 5 min, and the products were analyzed by 10% SDS-PAGE. The gel was dried and visualized by autoradiography.

Flow cytometry

As previously described (18), the cells were seeded onto 100-mm dishes and grown in DMEM supplemented with 10% FBS. After the cells had grown to subconfluence, they were rendered quiescent and chal-lenged with 10% FBS. Then after release using trypsin-EDTA, they were harvested at various times, washed twice with PBS/0.1% dextrose, and fixed in 70% ethanol at 4 C. Nuclear DNA was stained with a reagent containing propidium iodine (50 mg ml⫺1) and DNase-free RNase (2 U ml⫺1) and measured using a fluorescence-activated cell sorter (FACS). The proportion of nuclei in each phase of the cell cycle was determined using established CellFIT DNA analysis software (Becton Dickinson and Co., San Jose, CA).

Statistical analysis

Values represent the means⫾ sem. Three to six samples were ana-lyzed in each experiment. Comparisons were subjected to one-way ANOVA followed by Fisher’s least significant difference test. Signifi-cance was accepted at P⬍ 0.05.

Results

Arrest of cell cycle in G0/G1

Previously we demonstrated that progesterone inhibited DNA synthesis and decreased cell number in cultured RASMCs in a dose-dependent manner (14). To further study the actions of progesterone on the cell cycle, the cells were switched to media with 0.04% FBS for 72 h to render them quiescent and synchronize their cell cycle activities. Then they were returned to media with 10% FBS and, at various times thereafter, they were treated with [3H]thymidine.

Fig-ure 1A shows a reduction of the thymidine incorporation into RASMCs during the S-phase of the cell cycle. Figure 1B shows the FACS analyses of DNA content at 24 h after release from quiescence by incubation in culture media supple-mented with 10% FBS and PBS or progesterone (500 nm) in PBS. The data reveal that progesterone induced a significant accumulation of cells at the G0/G1 phase of the cell cycle, suggesting that the observed growth inhibition effect of

(4)

gesterone was due to a retardation of DNA replication, thereby inhibiting further progress in the cell cycle.

Alterations in cell cycle activity

It has been generally believed that coordinated successive activation of certain CDKs occurs late in the G1 phase and is instrumental in the transition from the G1 to the S-phase. This CDK activation is in turn modulated by association with a number of regulatory subunits called cyclins and a group of CDK-inhibitory proteins designated CKIs. Cyclins have been identified as cyclins A, D1, D3, and E. Using Northern blot analysis, we previously demonstrated that progesterone dose dependently inhibits the expression levels of cyclin A and E mRNA but not cyclin B and D1 mRNA (14). As illus-trated in Fig. 2, Western blot analysis demonsillus-trated that progesterone dose dependently inhibited the protein levels of cyclin A and E but not cyclin D1. We also examined the changes of CDK levels in the progesterone-treated RASMCs. In response to progesterone treatment, the levels of CDK2, but not CDK4, protein were decreased in a dose-dependent manner (Fig. 2). Because not only the protein levels of cyclins and CDKs but also a group of CKIs can control the CDK activity, we examined the protein levels of p21 and p27, two

known CKIs, in the progesterone-treated RASMCs. Figure 3A shows that the protein levels of p21 and p27 were in-creased in the progesterone-treated RASMCs, compared with the PBS-treated cells (control). We further conducted an immunoprecipitation assay to examine the effect of proges-terone on the formation of CDK-CKI complex. In progest-erone-treated cells, the formations of the CDK2-p21 and CDK2-p27 complex, but not CDK4-p21 and CDK4-p27 com-plex, were increased (Fig. 3B), and the assayable CDK2 ki-nase activity, but not CDK4 activity, was decreased (Fig. 3C). These changes are in a dose-dependent manner. These find-ings suggest that progesterone induced an inhibition of CDK2 activity and led to the impairment of RASMCs in the transition from G1 to S-phases.

P21 and p27 are the key regulators for the progesterone-induced G0/G1 arrest

As illustrated in Fig. 3, A and B, the levels of p21 and p27 protein and formations of CDK2-p21 and CDK2-p27 complex were dose dependently increased in progesterone-treated RASMCs, suggesting that up-regulation of p21 and p27 might be responsible for the progesterone-mediated G0/G1 arrest in these cells. To further demonstrate that in the

FIG. 1. Time-dependent inhibition of cell cycle in RASMCs by progesterone. To study the time-dependent progester-one on the cell cycle, [3H]thymidine

in-corporation was conducted after RASMC release from quiescence by incubation in culture media supple-mented with 10% FBS and PBS (con-trol) or 500 nMprogesterone in PBS (A). Comparisons were subjected to ANOVA followed by Fisher’s least significant difference test. Significance was ac-cepted at P ⬍ 0.05. *, Progesterone-treated group different from PBS-treated group (n⫽ 6). FACS analysis of DNA content was performed after 24-h release from quiescence by incubation in culture media supplemented with 10% FBS and PBS without (control) or with 500 nMprogesterone (B). Percent-age of cells at the G0/G1, S, or G2/M phase of the cell cycle was determined using established CellFIT DNA analy-sis software. Four samples were ana-lyzed in each group, and values repre-sent the meanSEM.

(5)

progesterone-treated RASMCs, increased p21 and p27 ex-pressions correlated with G0/G1 arrest, the experiment il-lustrated in Fig. 4 was carried out. Thus, in the sample labeled P4 for progesterone (500 nm) treated alone, the [3

H]thymi-dine incorporation was decreased. Sample P4⫹AS p21 was treated with a 20-nm p21 antisense oligonucleotide (AS), which blocked the expression of p21, and sample P4⫹AS p27 was treated with a 20 nm p27 AS, which blocked the expres-sion of p27. Treatment of RASMC with AS p21 or AS p27 alone did not cause any significant change in [3H]thymidine

incorporation into RASMCs (data not shown). Consequently, pretreatment of the RASMCs with AS p21 or AS p27 partially reversed the progesterone-induced decrease in [3

H]thymi-dine incorporation. The progesterone-induced inhibition in [3H]thymidine incorporation of RASMCs was completely

reversed by a combined administration of both antisense oligonucleotides to p21 and p27 together (Fig. 4).

Discussion

Progesterone has been suggested to be a paradoxical hor-mone existing either growth stimulatory or inhibitory effects, depending on the tissue and treatment regimen (19 –23). For example, progesterone inhibits epithelial growth in the uterus (24). On the other hand, in animals with established progesterone receptor-positive mammary tumors, proges-terone usually stimulates proliferation (25). We previously demonstrated that progesterone, the natural hormone, at physiologic levels inhibited DNA synthesis and decreased cell number in cultured RASMCs in a dose-dependent man-ner. The specificity of progesterone’s inhibitory effect on [3H]thymidine incorporation was confirmed by

preincuba-tion of RASMCs with progesterone receptor antagonist RU486, which antagonized the inhibition of [3H]thymidine

incorporation induced by progesterone (14). In the present study, we conducted [3H]thymidine incorporation and flow

cytometry analyses to further demonstrate that progesterone at physiologic levels (5–500 nm) inhibited DNA synthesis in cultured RASMCs in a time-dependent manner and arrested the cells at the G0/G1 phase of the cell cycle (Fig. 1).

Observation of intracellular events associated with the progression of cell cycle activity have suggested that coor-dinated successive activation of certain CDKs occurs late in the G1 phase and is instrumental in the transition from the G1 to the S-phase (26, 27). This CDK activation is in turn modulated by association with a series of regulatory subunits called cyclins and with a group of CDK-inhibitory proteins designated CKIs (28). Cyclins have been identified as cyclins A, D1, D3, and E, whereas the most common CDKs are designated CDK2 and CDK4. Cyclin A-CDK2 and cyclin E-CDK2 complexes form late in the G1 phase as cells prepare to synthesize DNA (29), and formation of the cyclin E com-plex is a rate-limiting step in the G1/S transition (30). It has been suggested that progesterone-induced entry of cells into the S-phase of the cell cycle is accompanied by transient increases of cyclin D1 and CDK4 activity (20, 21). In contrast, progesterone-induced retardation of cell entering into the S-phase is accompanied by down-regulation of cyclins D, A, and B and sequential increases in the levels of the CDK inhibitors, p21 and p27 (21). A study done in T-47D breast cancer cells demonstrated that progestin-mediated growth arrest was preceded by inhibition of cyclin D1-CDK4, cyclin D3-CDK4, and cyclin E-CDK2 kinase activities and reduced phosphorylation of pRB and p107. This was accompanied by decreases in the expression of cyclins D1, D3, and E protein; decreased abundance of cyclin D1- and cyclin D3-CDK4 com-plexes; increased association of the CDK inhibitor, p27, with the remaining CDK4 complexes; and changes in the molec-ular masses and compositions of cyclin E complexes (23).

Previously we demonstrated that progesterone at a range of concentrations (5–500 nm) dose dependently decreased the levels of cyclin A and cyclin E mRNA (14). This finding led us to propose that progesterone inhibits arterial smooth mus-cle cell proliferation by interrupting the cell cymus-cle at the G1/S transition. In the present study, we further demonstrated that progesterone dose dependently decreased the levels of cyclin A, cyclin E, and CDK2 protein but not cyclin D1 and CDK4 protein. Moreover, treatment of RASMCs with progesterone resulted in an increase in the levels of p21 and p27 protein at 24 h after treatment. In accord with the established notion that p21 and p27 are two known CDK inhibitors, we found in progesterone-treated cells that the formations of the CDK2-p21 and CDK2-p27complex were increased, and the assayable CDK2 kinase activity was decreased. In contrast, the formations of the CDK4-p21 and CDK4-p27 complex and the assayable CDK4 kinase activity were not changed sig-nificantly. The progesterone effect on the CDK activity ap-pears to be tissue specific and more analogous to the effects on the uterine epithelium than on breast cancer cells. Pre-treatment of uterine epithelial cells with progesterone abro-gated estrogen-induced cyclin E-CDK2 activation (31). In breast cancer cells, on the other hand, the progestin-induced

FIG. 2. Effect of progesterone on the protein levels of cyclins and CDKs. Proteins were extracted from the cultured RASMCs at 24 h after progesterone treatment and probed with proper dilutions of specific antibodies. Progesterone dose dependently decreased the lev-els of cyclin A and cyclin E protein as well as CDK2 protein but not cyclin D1 and CDK4 protein. Membrane was probed with anti-G3PDH antibody to verify equivalent loading.

(6)

growth inhibition was preceded by inhibition of cyclin D1-Cdk4, cyclin D3-D1-Cdk4, and cyclin E-Cdk2 kinase activities (19, 23, 32).

Previously, it has been demonstrated that regulation of transcription of the p21 promoter is involved in the proges-terone-induced cell cycle arrest at the transition of the cells from the G1 phase to the S-phase (33). The important role of p21 and p27 play on the progesterone-induced inhibition of DNA synthesis was further confirmed by the demonstration that pretreatment of the RASMCs with a p21 or p27 antisense oligonucleotide reduced the progesterone-induced inhibi-tion of [3H]thymidine incorporation into RASMCs.

More-over, a combined treatment of RASMC with p21 and p27 antisense oligonucleotides completely reversed the proges-terone-induced inhibition of [3H]thymidine incorporation.

Accordingly, we concluded that progesterone may inhibit RASMCs proliferation by increasing the levels of p21 and p27 protein, which in turn inhibit CDK2 kinase activity, and finally impair the transition of the cells from the G1 phase to the S phase.

In conclusion, the results from the present study indicate that progesterone-induced cell cycle arrest in RASMCs oc-curred when the cyclin-CDK system was inhibited just as p21 and p27 protein levels were augmented. Although animal

FIG. 3. Effect of progesterone on levels of CKI protein, CKI-CDK association, and CDK kinase activity. A, Progester-one dose dependently increases the lev-els of p21 and p27 protein in RASMCs. Membrane was probed with anti-G3PDH antibody to verify equivalent loading. B, Treatment of RASMCs with progesterone (5–500 nM) induced up-regulation of the formations of CDK2-p21 and CDK2-p27 complex in a dose-dependent manner. The formations of CDK4-p21 and CDK4-p27 complex were not affected by progesterone treat-ment. CDK2 was immunoprecipitated by anti-CDK2 antibody, and CDK2-p21 complex was detected by p21 anti-body, whereas CDK2-p27 complex was detected by anti-p27 antibody. CDK4 was immunoprecipitated by anti-CDK4 antibody, and CDK4-p21 complex was detected by anti-p21 antibody, whereas CDK4-p27 complex was detected by anti-p27 antibody. C, The CDK2 kinase activity was decreased dose depen-dently by progesterone treatment, whereas CDK4 kinase activity was not significantly changed. Results from a representative experiment are shown. Mean values of CDK2 and CDK4 en-zyme activity from three experiments are shown in the parentheses (means SEM). The CDK2 and CDK4 kinase ac-tivity were determined as described in Materials and Methods.

(7)

studies of progesterone-mediated antiatherosclerosis are still ongoing, the findings from the present studies suggest the potential applications of progesterone in the treatment of atherosclerosis.

Acknowledgments

Received January 9, 2003. Accepted March 31, 2003.

Address all correspondence and requests for reprints to: Wen-Sen Lee, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110, Taiwan. E-mail: wslee@tmu.edu.tw.

This work was supported by research grants from the National Science Council of the Republic of China (TMC86-Y05-B111, NSC88-2314-B-038-121).

References

1. Libby P, Hansson GK 1991 Involvement of the immune system in human atherogenesis: current knowledge and unanswered questions. Lab Invest 64: 5–15

2. Munro JM, Cotran RS The pathogenesis of atherosclerosis: atherogenesis and inflammation 1988 Lab Invest 58:249 –261

3. Ross R 1993 The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature 362:801– 809

4. Schwartz SM, Heimark RL, Majesky MW 1990 Developmental mechanisms underlying pathology of arteries. Physiol Rev 70:1177–1209

5. Furman RH 1968 Are gonadal hormones (estrogens and androgens) of sig-nificance in the development of ischemic heart disease. Ann NY Sci 149:822– 833

6. Stampfer MJ, Colditz GA 1991 Estrogen replacement therapy and coronary

heart disease: a quantitative assessment of the epidemiologic evidence. Prev Med 20:47– 63

7. Adams MR, Kaplan JR, Manuck SB, Koritnik DR, Parks JS, Wolfe MS,

Clarkson TB1990 Inhibition of coronary artery atherosclerosis by 17-␤

estra-diol in ovariectomized monkeys. Lack of an effect of added progesterone. Arteriosclerosis 10:1051–1057

8. Haarbo J, Leth-Espensen P, Stender S, Christiansen C 1991 Estrogen mono-therapy and combined estrogen-progesterone replacement mono-therapy attenuate aortic accumulation of cholesterol in ovariectomized cholesterol-fed rabbits. J Clin Invest 87:1274 –1279

9. Foegh ML, Asotra S, Howell MH, Ramwell PW 1994 Estradiol inhibition of arterial neointimal hyperplasia after balloon injury. J Vasc Surg 19:722–726 10. Sullivan Jr TR, Karas RH, Aronovitz M, Faller GT, Ziar JP, Smith JJ,

O’Donell Jr TF, Mendelsohn ME1995 Estrogen inhibits the response-to-injury in a mouse carotid artery model. J Clin Invest 96:2482–2488

11. Grodstein F, Stamper M 1995 The epidemiology of coronary heart disease and estrogen replacement in postmenopausal women. Prog Cardiovasc Dis 38: 199 –210

12. Rekhter MD, Gordon D 1995 Active proliferation of different cell types, including lymphocytes, in human atherosclerotic plaques. Am J Pathol 147: 668 – 677

13. Barr DP, Russ EM, Eder HA 1952 Influence of estrogens on lipoproteins in atherosclerosis. Trans Assoc Am Physicians 65:102–113

14. Lee WS, Harder JA, Yoshizumi M, Lee ME, Haber E 1997 Progesterone inhibits arterial smooth muscle cell proliferation. Nat Med 3:1005–1008 15. Jain M, He Q, Lee WS, Kashiki S, Foster LC, Tsai JC, Lee ME, Haber E 1996

Role of CD44 in the reaction of vascular smooth muscle cells to arterial wall injury. J Clin Invest 97:596 – 603

16. Lin SY, Liu JD, Chang HC, Yeh SD, Lin CH, Lee WS 2002 Magnolol sup-presses proliferation of cultured human colon and liver cancer cells by inhib-iting DNA synthesis and activating apoptosis. J Cell Biochem 84:532–544 17. Lin SY, Liang YC, Ho YS, Tsai SH, Pan S, Lee WS 2002 Involvement of both

extracellular signal-regulated kinase and c-jun N-terminal kinase pathways in the 12-o-tetradecanoylphorbol-13-acetate-induced upregulation of p21Cip1in

colon cancer cells. Mol Carcinog 35:21–28

18. Lin SY, Chang YT, Liu JD, Yu CH, Ho YS, Lee WS 2001 Molecular mechanisms of apoptosis induced by Magnolol in colon and liver cancer cells. Mol Carcinog 32:73– 83

19. Musgrove EA, Lee CS, Sutherland RL 1991 Progestins both stimulate and inhibit breast cancer cell cycle progression while increasing expression of transforming growth factor␣, epidermal growth factor, c-fos, and c-myc genes. Mol Cell Biol 11:5032–5043

20. Musgrove EA, Hamilton JA, Lee CS, Sweeney KJ, Watts CK, Sutherland RL 1993 Growth factor, steroid, and steroid antagonist regulation of cyclin gene expression associated with changes in T-47D human breast cancer cell cycle progression. Mol Cell Biol 13:3577–3587

21. Groshong SD, Owen GI, Grimison B, Schauer IE, Todd MC, Langan TA,

Sclafani RA, Lange CA, Horwitz KB1997 Biphasic regulation of breast cancer cell growth by progesterone: role of the cyclin-dependent kinase inhibitors, p21 and p27(Kip1). Mol Endocrinol 11:1593–1607

22. Graham JD, Clarke CL 1997 Physiological action of progesterone in target tissues. Endocr Rev 18:4502– 4519

23. Musgrove EA, Swarbrick A, Lee CS, Cornish AL, Sutherland RL 1998 Mech-anisms of cyclin-dependent kinase inactivation by progestins. Mol Cell Biol 18:1812–1825

24. Clarke CL, Sutherland RL 1990 Progestin regulation of cellular proliferation. Endocr Rev 11:266 –301

25. Horwitz KB 1992 The molecular biology of RU486. Is there a role for anti-progestins in the treatment of breast cancer? Endocr Rev 13:146 –163 26. Hunter T, Pines J 1994 Cyclins and cancer: cyclin D and CDK inhibitors come

of age. Cell 79:573–582

27. Morgan DO 1995 Principles of CDK regulation. Nature 374:131–134 28. Sherr CJ, Roberts JM 1995 Inhibitors of mammalian G1 cyclin-dependent

kinases. Genes Dev 9:1149 –1163

29. Lees E 1995 Cyclin dependent kinase regulation. Curr Opin Cell Biol 7:773–780 30. Sherr CJ 1993 Mammalian G1 cyclins. Cell 73:1059 –1065

31. Tong W, Pollard JW 1999 Progesterone inhibits estrogen-induced cyclin D1 and cdk4 nuclear translocation, cyclin E- and cyclin A-cdk2 kinase activation, and cell proliferation in uterine epithelial cells in mice. Mol Cell Biol 19:2251– 2264

32. Swarbrick A, Lee CS, Sutherland RL, Musgrove EA 2000 Cooperation of p27(Kip1) and p18(INK4c) in progestin-mediated cell cycle arrest in T-47D breast cancer cells. Mol Cell Biol 20:2581–2591

33. Owen GI, Richer JK, Tung L, Takimoto G, Horwitz KB 1998 Progesterone regulates transcription of the p21WAF1cyclin-dependent kinase inhibitor gene

through Sp1 and CBP/p300. J Biol Chem 273:10696 –10701

FIG. 4. Involvement of p21 and p27 in the progesterone-induced decrease of [3H]thymidine incorporation. Antisense p21 or p27

oligo-nucleotide alone partially reversed the progesterone-mediated decrease of [3H]thymidine incorporation. However, the

progesterone-induced inhibition in [3H]thymidine incorporation of RASMCs was

completely reversed by a combined administration of both antisense oligonucleotides to p21 and p27 together. AS p21 or p27 was added to RASMCs at a final concentration up to 20 nMat 1 h before the cell was challenged with 2% FBS, and 500 nMprogesterone for an additional 24 h. [3H]Thymidine incorporation was conducted after RASMC

re-lease from quiescence by incubation in culture media supplemented with 2% FBS and PBS (control) or 500 nMprogesterone in PBS. Three to four samples were analyzed in each group, and values represent the means⫾SEM. P4, Progesterone; AS p21, antisense p21 oligonucleo-tide; AS p27, antisense p27 oligonucleotide.

參考文獻

相關文件

The functional fall of the salivary secretion observed at 180 days post irradiation was not only associated with a reduction of gland mass but also to an alteration of the

The functional fall of the salivary secretion observed at 180 days post irradiation was not only associated with a reduction of gland mass but also to an alteration of the

 Finally, the immunohistochemical results in the present case highlights that Bcl-2 negativity in granular cells indicating an apoptotic process, CD-68 positivity in granular

(E) Varying numbers of tumour cells expressing cancer antigen 125 are present in the lining of tubules (objective magnification ¥10).. The arrowed cells show strong nuclear

Its main tool is the stem cells that are seeded on the surface of biomaterials (scaffolds), in order to create a biocom- plex. Several populations of mesenchymal stem cells are found

(1) Skin melanoma cells show molecular alterations of the RAS-BRAF-MEK-ERK mitogen activated protein kinase (MAPK) signalling pathway, mediating uncontrolled proliferation of

Giant cells are also seen in other diseases like giant cell granuloma of the jaws, traumatic bone cyst, aneurysmal bone cyst, and jaw tumor of

stained negative for a-smooth muscle actin because the main body of the tumor consisted of epithelioid or epithe- lial cells that contained few positive smooth muscle cells.. We