Вы находитесь на странице: 1из 6

Bone 32 (2003) 136 141 www.elsevier.

com/locate/bone

The effects of estrogen on osteoprotegerin, RANKL, and estrogen


receptor expression in human osteoblasts
S. Bord,* D.C. Ireland, S.R. Beavan, and J.E. Compston
University of Cambridge School of Clinical Medicine, Addenbrookes Hospital, Box 157, Cambridge CB2 2QQ, UK
Received 28 February 2002; revised 28 August 2002; accepted 8 October 2002

Abstract
Estrogen is essential for bone growth and development and for the maintenance of bone health in adulthood. The cellular responses of
osteoblasts and osteoclasts to estrogen are initiated via two high-affinity receptors (ERs). Osteoblasts synthesize RANKL (receptor activator
of NF-B ligand), necessary for osteoclast formation and function, and osteoprotegerin (OPG), its decoy receptor. To investigate the effects
of estrogen on the expression of OPG, RANKL, and ERs in human osteoblasts, cells were cultured with physiological (1010 M) and
high-dose (107 M) 17-estradiol for 24 and 48 h. Proteins and corresponding mRNA levels were quantitatively determined by
immunocytochemistry and RTPCR. OPG expression was significantly increased three- and sevenfold at 24 h with 1010 M (P 0.05)
and 107 M (P 0.01) estradiol, respectively, compared to untreated cells. Similar but smaller increases were seen at 48 h (P 0.05).
Osteoblasts treated with estradiol demonstrated increased RANKL protein expression at 24 h (P 0.05), but this was not maintained at 48 h.
ER expression was significantly increased by high-dose estradiol (P 0.01) at 24 h and dose-dependently increased at 48 h (P 0.01),
while ER was only increased at 24 h (P 0.01). The estrogen-induced protein expression of ER, OPG, and RANKL was abrogated when
cells were cultured in the presence of the estrogen antagonist ICI 182,780. mRNA levels at 24 h demonstrated a significant suppression of
RANKL with the low-dose but not the high dose. ER mRNA but not ER expression was up-regulated by estrogen. Our results suggest
that estrogen may exert its anti-resorptive effects on bone, at least in part, by stimulating ER and OPG expression in osteoblasts.
2003 Elsevier Science (USA). All rights reserved.

Keywords: Estrogen; Osteoblasts; Osteoprotegerin; RANKL; Estrogen receptors

Introduction mone receptor and a member of a family of activated tran-


scription factors that can initiate or enhance the transcrip-
It is well established that estrogen has multifunctional tion of genes. Two ER subtypes have been identified, and
roles influencing growth, differentiation, and function in . They are distinct proteins encoded by separate genes and
many tissues. It is an important factor in the maintenance of located on different chromosomes.
bone health, estrogen deficiency at the time of menopause RANKL (receptor activator of NF-B ligand), a mem-
being associated with bone loss. Administration of conven- brane-bound molecule, is a newly identified member of the
tional doses of hormone replacement therapy prevents tumor necrosis factor (TNF) ligand family and has been
menopausal bone loss by reducing bone turnover and inhib- shown to be crucial for osteoclast formation [6]. Two re-
iting osteoclast activity [1], whereas high doses of estrogen ceptors for RANKL have been identified, RANK, a mem-
have been shown to exert anabolic skeletal effects in rodents brane-bound signaling receptor expressed on the cell sur-
and postmenopausal women [25]. Estrogens diffuse in and
face of osteoclast progenitors, and osteoprotegerin (OPG), a
out of cells, but are retained in target cell nuclei by the
secreted cytokine receptor. OPG, a member of the tumor
estrogen receptor protein (ER). The ER is a nuclear hor-
necrosis factor receptor (TNF-R) superfamily [8,9] acts as a
decoy receptor by blocking the interaction of RANKL with
* Corresponding author. Fax: 44-0-1223-336846. its functional receptor RANK [10], thereby inhibiting oste-
E-mail address: sb201@cam.ac.uk (S. Bord). oclastogenesis.

8756-3282/03/$ see front matter 2003 Elsevier Science (USA). All rights reserved.
doi:10.1016/S8756-3282(02)00953-5
S. Bord et al. / Bone 32 (2003) 136 141 137

In vitro studies have demonstrated the synthesis of OPG moved by washing in PBS prior to addition of a biotinylated
by stromal cell lines [11] and osteoblasts [12] and its stim- secondary antibody (horse antirabbit or horse antigoat, Vec-
ulation by estrogen [13]. Udagawa et al. [14] reported that tor Laboratories, at 3.5 g/ml) for 30 min at room temper-
osteoblasts constitutively express RANKL mRNA, the bone ature. Following washings with PBS to remove unbound
resorbing capacity of osteoclasts being enhanced when antibody the signal was amplified by the addition of an
cocultured with osteoblasts. avidin biotin complex (ABC) Elite substrate (Vector Lab-
The aim of this study was to investigate the relative oratories). Signal was detected using DAB (Vector Labora-
effects of estrogen on ER, OPG, and RANKL mRNA and tories). Cells were lightly counterstained with Gills Haema-
protein expression in human osteoblasts. toxylin (1:50, Sigma) for 1 min, blued in tap water for 10
min, and air-dried prior to mounting. The specificity of the
antibody reaction was ascertained by substituting the pri-
Materials and methods mary antibody with nonimmune serum at the same IgG
concentration and omission of primary and secondary anti-
Cell culture and immunolocalization bodies.

Primary human osteoblasts from two young female do- Quantitation of immunolocalization
nors (1 day and 1 year) were supplied by Clontech Labo-
ratories (Basingstoke, UK), with each grown to confluence Cells were examined by light microscopy with a Nikon
and expanded to yield cells for three replica experiments. E-800 fitted with a Basler digital camera. The intensity and
Cells were seeded into eight-well chamber slides (NUNC) extent of staining were measured using Lucia G image
at 104 cells/well in McCoys 5A medium supplemented with analysis. Thresholds were set to detect only positive stain-
10% heat-inactivated FBS (Life Technologies), penicillin/ ing with five fields of view examined for each antibody for
streptomycin (Life Technologies), and ascorbic acid (100 each experiment. Experiments were repeated three times.
mM, Wako, Alpha Labs, Eastleigh, UK). Cultures were Results in each group are expressed as the mean relative
incubated at 37 C in a humidified chamber with 5% CO2. change in the E-treated cells compared to untreated cells
After 24 h the medium was removed and replaced with (SD).
medium minus or plus 17-estradiol (E2, Sigma), at phys-
iological E2 (low dose, 1010 M) or a saturating concentra- Cell culture for RNA determination and relative
tion of E2 (high dose, 107 M) for 24 or 48 h. Similar quantification of gene expression
cultures were treated with the estrogen antagonist ICI 182,
780 (108 M, Tocris Cookson) for 24 h. Cells (as above) were plated at 105 cells/ml and cultured
At the end of the incubation period (24 or 48 h) the in T 75 flasks in medium (as above) for 24 h. The medium
medium was removed and cells were rinsed in phosphate- was removed and replaced with fresh medium containing no
buffered saline (PBS), pH 7.4, fixed with 4% paraformal- E2, low-dose E2, or high-dose E2 as above for 24 h. Cells
dehyde for 5 min at room temperature, and immunostained were removed with TRIzol and RNA was extracted accord-
using an indirect immunoperoxidase method. Following fix- ing to manufacturers instructions (Life Technologies, Inc.,
ation, cells were washed with PBS and endogenous perox- Paisley, UK).
idase activity was blocked with ImmunoPure Suppressor To prepare RNA standards PCR products for ER, ER,
(Pierce) for 30 min at room temperature. Further PBS OPG, RANKL, and GAPDH were made by RTPCR with
washes were followed by blocking with 15% normal horse primer sequences shown in Table 1. PCR products were
serum (Vector Laboratories, Peterborough, UK) for 30 min cloned into the T-tailed vector pCR II-TOPO (Invitrogen),
at room temperature. Primary antibodies to ER (1.0 g/ml, which has opposing SP6 and T7 RNA polymerase promotor
rabbit polyclonal HC-20 mapping to the carboxy terminus sites. BamHI restriction sites were added to forward primers
of ER and shown not to cross-react with ER) and ER so that the orientation of each insert could be determined by
(1.0 g/ml, rabbit polyclonal H-150 raised against a recom- cutting the plasmids with BamHI. Plasmids containing in-
binant protein corresponding to amino acids 1150 mapping serts suitable for transcription with SP6 RNA polymerase
at the amino terminus of human ER and shown to be were selected and cut with NotI. RNA transcripts were
non-cross-reactive with ER), OPG (1.0 g/ml, goat poly- purified using Microspin S-300 columns (Pharmacia) after
clonal N-20 raised against a peptide mapping the amino treatment with DNase to remove plasmid DNA. mRNA
terminus of human OPG), and RANKL (1.0 g/ml, goat levels were determined using one-step RTPCR reagents
polyclonal C-20 raised against a peptide mapping at the (Applied Biosystems) in a Gene Amp 5700 SDS with
carboxy terminus of human RANKL) were obtained from Primer Express software used to design primers and probes.
Santa Cruz Biotechnology (Santa Cruz, CA). These anti- Primers pairs were chosen to include introns in the gene
bodies, diluted in 0.1% bovine serum albumin (Vector Lab- sequence and probes to span intron exon boundaries (Table
oratories), were added and incubated overnight at 4 C in a 2). A standard curve was included in each assay so that the
humidified chamber. Unbound primary antibody was re- overall efficiency of the assay could be calculated. mRNA
138 S. Bord et al. / Bone 32 (2003) 136 141

Table 1
Sequences of cloning primers

Gene Forward primer Reverse primer

GAPDH TGAAGGTCGGAGTCAACGGATTTG GTTGGTGGTGCAGGAGGCATTGCT


ER* AATTCAGATAATCGACGCCAG GTGTTTCAACATTCTCCCTCCTC
ER* TAGTGGTCCATCGCCAGTTAT GGGAGCCACACTTCACCAT
OPG GTCAAGCAGGAGTGCAATCG GAGTTGATTCACTGTTTCCGG
RANKL ATCCCACTCGGTTCCCATA CCCTGACCAATACTTGGTGC

* Arts et al. (1997) [20].

levels were quantified using the comparative threshold-cy- P 0.01). At 48 h there was a more modest but still
cle (CT) method [7]. First the amount of target mRNA in significant increase in OPG expression in the estrogen-
each sample was normalized to the amount of housekeeper treated cells (low dose, 1.2 0.09, high dose, 1.3 0.16,
mRNA (GAPDH), designated as calibrator, to give CT (CT P 0.05) (Fig. 1). RANKL expression was elevated in the
target CTGAPDH). Second, the amounts of target mRNA in presence of E2 at 24 h but not at 48 h. At 24 h there was a
the samples were compared using the formula Amount of 2.35-fold (0.74, P 0.05) increase with low-dose E2 and
target mRNA 2CT, where CT CTsample1 a 2.1-fold (0.67, P 0.05) increase with high-dose E2
CTsample2 (calibrator), assuming that the efficiencies of the compared to untreated cells (Fig. 1). ER expression varied
PCR reaction were close to 1. The efficiency of each assay with time and dose of E2 treatment. At 24 h high-dose E2
was calculated using the formula E 101/S 1, where S elicited a 2.05-fold (0.11, P 0.01) increase in ER ex-
is the slope of the standard curve. pression but there was no change in the low-dose E2-treated
Four replicates were performed for each experimental cells. At 48 h a dose-dependent increase was observed (low
point and experiments repeated three times. The results dose, 2.14 0.44, high-dose, 2.57 0.68, P 0.01) (Fig. 1).
shown (Fig. 3) are of a representative experiment. ER exhibited a significant dose-dependent increase at 24 h
(low dose, 1.72 0.20, high dose, 2.05 0.44, P 0.01) but
Statistical analysis had returned to basal levels at 48 h (Fig. 1).
Cells cocultured with E2 and the estrogen antagonist ICI
Statistical analysis for protein and mRNA data was per- 182,780 showed no estrogen-induced increase in OPG,
formed using the approximate test for unequal variance RANKL, or ER expression. In all cultures the expression
based on the t distribution [15]. levels remained at basal levels (Fig. 2).

mRNA expression
Results
mRNA data were collected at 24 h to determine that
Protein expression changes in protein levels were as a result of changes in
transcription rather than proteasome-mediated receptor deg-
The result for untreated cells in each group was given a radation or translation. The amount of target mRNA for
value of 1, and data for treated cells in each group were each sample was normalized to the amount of the house-
expressed as a relative ratio. OPG protein expression mea- keeper mRNA (GAPDH). GAPDH expression did not
sured at 24 h demonstrated a significant 3-fold increase with change significantly with estradiol treatment. Approximate
low-dose E2 compared to untreated cells (3.00 1.05, P relative levels of mRNA expression with GAPDH as 1 were
0.05) and a 7-fold increase with high-dose E2 (6.9 1.80, OPG, 0.25; ER, 0.00025; ER, 0.00004; and RANKL,

Table 2
GeneAmp 5700 SDS primers and probes

Gene Forward primer Reverse primer Probe

GAPDH TTTTAACTCTGGTAAAGTGGATATTGTTG TGACGGTGCCATGGAATTT ATTGACCTCAACTACATGGTTTACA


TGTTCCAATATG
ER TGCTTCAGGCTACCATTATGGA GTTTTTATCAATGGTGCACTCGTA ACACATATAGTCGTTATGTCCTTG
AATACTTCTCTTGAAGAA
ER TCAAAAGAGTCCCTGGTGTGAAG CTCTTTGAACCTGGACCAGTAACAG CGGTTCCCACTAACCTTCCTTTTC
AGTGTCTCT
OPG GAGATAGAGTTCTGCTTGAAACA CCATCTGGACATCTTTTGCAAA TGCAAGCTGGAACCCCAGAGCG
RANKL CAGCCTTTTGCTCATCTCACTATTAA GGTACCAAGAGGACAGACTCACTTTA ACCGACATCCCATCTGGTTCCC
S. Bord et al. / Bone 32 (2003) 136 141 139

Fig. 1. Immunohistochemistry of osteoprotegerin (OPG), RANKL, and estrogen receptor (ER) and protein expression by osteoblasts at 24 and 48 h was
quantitatively measured by image analysis. Results from untreated cells were given a value of 1 and estrogen-treated cells a relative ratio. Results SD, *
P 0.05, ** P 0.01, compared to untreated cells.

0.00002. To compare changes in expression levels with osteoblasts [16 18] and Udagawa et al. [14] reported the
treatment, the result for untreated cells in each group was constitutive expression of RANKL by osteoblasts. In addi-
given a value of 1, and data for treated cells in each group tion, various reports show the induction of OPG by estrogen
were expressed as a relative ratio. At 24 h RANKL mRNA [13,19]. However, to our knowledge, this is the first report
expression was suppressed by low-dose E2 (P 0.05) but of estrogen-induced changes in OPG, RANKL, and ER
not by by high-dose E2. OPG, ER, and ER mRNA expression measured in the same human osteoblast cultures.
expressions were unchanged at 24 h with high-dose E2 The use of image analysis in our study provided quantitative
treatment. With low-dose E2 treatment only ER was in- data for protein synthesis that adds to the observational
creased (P 0.05) (Fig. 3). reports in other studies.
We have previously reported the presence of both ER
and ER in human osteoblastic cells in vitro and in devel-
Discussion oping human bone in vivo [16]. The current study demon-
strates that in vitro osteoblastic cells respond initially to
This in vitro study demonstrated that estrogen elicits estrogen by increasing ER and protein expression. By
changes in cellular protein and mRNA expression in human 48 h ER expression had returned to basal levels, whereas
osteoblastic cells with OPG and ER protein expression ER expression continued to rise. From this study it was not
significantly increased. The estrogen-induced changes in possible to establish the time point of maximal mRNA
ER expression and up-regulation of the decoy receptor OPG expression or at which time point this was reflected by
suggest that osteoblasts may be involved in the mediation of maximal protein expression. However, the mRNA data at
the estrogen-induced anabolic skeletal effects in bone. We 24 h were generally in concordance with the 48-h protein
and other groups have demonstrated the presence of ERs in expression. ER showed a significant increase at 24 h,
which agreed with the protein data at 48 h, while ER

Fig. 2. Immunohistochemistry of OPG, RANKL, ER and ER protein


expression by osteoblasts at 24 h with and without the estrogen antagonist
ICI 182,780 was quantitatively measured by image analysis. Results from Fig. 3. OPG, RANKL, ER, and ER mRNA expression by osteoblasts
untreated cells were given a value of 1 and estrogen-treated cells a relative was measured at 24 h. Results were normalized to GAPDH. Results from
ratio. Results SD, * P 0.05, ** P 0.01, compared to untreated untreated cells were given a value of 1 and estrogen-treated cells a relative
cells. ratio. Results SD, * P 0.05, compared to untreated cells.
140 S. Bord et al. / Bone 32 (2003) 136 141

mRNA demonstrated no change, which was reflected by the fected with ER, showed that 17-estradiol induced a dose-
protein expression at 48 h. Although the results for OPG and dependent increase in OPG protein secretion after 24 h of
ER demonstrated the same trends as the protein data these culture. This increase was completely abrogated by cocul-
results were not significant due to the wide variability be- ture with the estrogen antagonist ICI 182,780. They also
tween samples. demonstrated that the estrogen-induced effect was specific
Studies of human breast cancer tissue have shown that to 17-estradiol, as the estrogen isomer 17-estradiol had
ER-negative cells are more proliferative and less differen- no stimulatory effect. Using a primary cell line from young
tiated than ER-positive cells. In the ER-positive tumors human donors we have demonstrated similar effects to the
the amount of ER mRNA was correlated to age and post- transfected cells used by Hofbauer et al. [12,13]. We quan-
menopausal status, higher levels being demonstrated in el- tified cellular synthesis of OPG in response to estrogen and
derly women [11]. In studies conducted in our laboratory we show that protein expression is significantly increased in
have found that proliferating osteoblastic cells from young
estrogen-treated cells at both 24 and 48 h. However, OPG
donors do not show increasing ER/ER with time in
synthesis was measured only in osteoblasts and it is possible
culture, unlike cells from older donors, which mineralize
that levels of secreted OPG in the culture medium may
more rapidly and express increasing ER mRNA [17].
differ between treated and untreated cultures. Further work
Consistent with this, Arts et al. [20], using a mineralizing
is required to determine this fact. RANKL is a functional
immortalized osteoblast-like cell line, showed differential
mRNA expression of ER and ER during cell differenti- protein when expressed at the cell membrane. In our study
ation. ER expression was found to increase during culture, intense staining was seen on the cell surface of many os-
whereas ER levels, after an initial increase, stayed con- teoblasts and image analysis detection thresholds were set to
stant. In our current study, using young donor osteoblasts, identify these areas of dark staining. However, some intra-
ER continued to rise, while ER had returned to basal cellular intense staining was observed. As this might be
values at 48 h, indicating that the cells had not reached the representative of RANKL trafficking to the cell surface
mineralizing stage and collagen synthesis was not down- these measurements were retained.
regulated. Together these reports suggest a potential mech- Both doses of estrogen produced an increase in RANKL
anism for the observed anabolic effect produced by high- protein synthesis at 24 h. As estrogen is known to exert an
dose estrogen, with the ER/ER ratio dependent on the anti-resorptive effect on bone this finding was unexpected.
age of the person, the dose, and the length of time of However, protein levels had returned to basal values by
estrogen treatment. However, our complete understanding 48 h, leaving only elevated OPG. Thus, in this system,
of the mechanisms of regulation of ERs has yet to be estrogen could alter the OPG/RANKL ratio against oste-
established. A recent study by Waters and colleagues [21] oclastogenesis.
using osteoblast cell lines stably expressing either ER or Interestingly in a study by Collin-Osdoby and colleagues
ER identified responses to estrogen that were both ER [25] increases in RANKL and OPG mRNA expression were
isoform-specific and differentiation stage-dependent, which seen in endothelial cells following an inflammatory stimu-
is consistent with reports from Ireland et al. [17]. Bonnelye lus. The RANKL/OPG ratio increased because OPG levels
and Aubin [22] have shown that the estrogen receptor- declined. In the same study experiments performed on
related receptor is codistributed with ER and ER in HBMSC cultured in the presence of PTH and vitamin D
different cohorts of osteoblastsic cells, raising the possibil- also showed a reciprocal expression pattern, with an early
ity that the receptors function in different pairs in different rise in OPG followed by a declined together with a delayed
groups of osteoblasts. These observations give rise to some
and sustained rise in RANKL. In contrast HMVEC treated
intriguing questions.
with these calciotrophic agents showed no change in their
RANKL, an essential factor for osteoclast formation and
expression patterns, indicting that different stimuli may
activity, and OPG are regulated by various hormones, cy-
affect the OPG/RANKL ratio in different ways and in dif-
tokines, and mesenchymal transcription factors. However,
OPG, which acts as a soluble neutralizing receptor to ferent cell types.
RANKL, is produced by cells of the osteoblastic lineage and To investigate the mechanism by which estrogen may
therefore likely to be a major regulator of bone metabolism. induce protein expression, osteoblasts were cultured with
Severe osteoporosis develops in mice with OPG gene dele- estradiol in the presence or the absence of the estrogen
tion [23], whereas mice overexpressing OPG develop os- antagonist ICI 182,780. The low- and high-dose estrogen-
teopetrosis [22]. Recently Croucher et al. [24] reported induced expression of OPG, RANKL, and ER was abro-
expression of RANKL in myeloma cells in mice. The in- gated by the estrogen antagonist, suggesting that the actions
crease in osteoclast number and the associated development of estrogen on these cytokines are mediated by ERs.
of bone disease, osteolytic lesions, and decrease in bone In summary, the data presented here suggest that es-
volume were prevented by treatment with recombinant trogen may exert its anti-resorptive effects on bone, at
OPG, indicating the importance of OPG in vivo. Hofbauer least in part, by stimulating ER and OPG expression in
et al. [13], using an osteoblast-like cell line stably trans- osteoblasts.
S. Bord et al. / Bone 32 (2003) 136 141 141

Acknowledgments ulated by vitamin D, bone morphogenetic protein and cytokines.


Biochem Biophys Res Commun 1998;250:776 81.
[13] Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Spelsberg TC,
This work was funded by the Wellcome Trust.
Riggs BL. Estrogen stimulates gene expression and protein produc-
tion of osteoprotegerin in human osteoblastic cells. Endocrinology
1999;140:436770.
References [14] Udagawa N, Takahashi N, Jimi E, Matsuzaki K, Tsurukai T, Itoh K,
Nakagawa N, Yasuda H, Goto M, Tsuda E, Higashio K, Gillespie
[1] Vedi S, Compston JE. The effects of long-term hormone replacement MT, Martin TJ, Suda T. Osteoblasts/stromal cells stimulate osteoclast
therapy on bone remodelling in postmenopausal women. Bone 1996; activation through expression of osteoclast differentiation factor/
19:5359. RANKL but not macrophage colony-stimulating factor. Bone 1999;
[2] Chow J, Tobias JH, Colston KW, Chambers TJ. Estrogen maintains 25:51723.
trabecular volume in rats not only by suppression of resorption but [15] Armitage P, Berry G. Statistical inference. In: Statistical methods in
also by stimulation of bone formation. J Clin Invest 1992;89:74 8. medical research, 3rd ed. Blackwell; 1994, p. 1157.
[3] Edwards MW, Bain SD, Bailey MC, Lantry MM, Howard GA. 17 [16] Bord S, Horner A, Beavan S, Compston J. Estrogen receptors and
Estradiol stimulation of endosteal bone formation in the ovariecto- are differentially expressed in developing human bone. J Clin
mised mouse: an animal model for the evaluation of bone-targeted Endocrinol Metab 2001;86:2309 14.
estrogens. Bone 1992;13:29 34. [17] Ireland DC, Bord S, Beavan SR, Compston JE. Effects of estrogen on
[4] Vedi S, Purdie DW, Ballard P, Bord S, Cooper AC, Compston JE. collagen synthesis by cultured human osteoblasts depend on the rate
Bone remodelling and structure in postmenopausal women treated of cellular differentiation. J Cell Biochem 2002;9999:17.
with long-term, high-dose estrogen therapy. Osteoporos Int 1999;10: [18] Onoe Y, Miyaura C, Ohta H, Nozawa S, Suda T. Expression of
52 8. estrogen receptor in rat bone. Endocrinology 1997;138:4509 12.
[5] Wahab M, Ballard P, Purdie DW, Cooper A, Wilson JC. The effect [19] Saika M, Inoue D, Kido S, Matsumoto T. 17-Estradiol stimulates
of long-term oestradiol implantation on bone mineral density in expression of osteoprotegerin by a mouse stromal cell line, ST-2, via
postmenopausal women who have undergone hysterectomy and bi- estrogen receptor-. Endocrinology 2001;142:220512.
lateral oophorectomy. Br J Obstet Gynaecol 1997;104:728 31. [20] Arts J, Kuiper GJM, Janssen JMMF, Gustafsson J-A, Lowik CWCM,
[6] Fuller K, Wong B, Choi Y, Chambers TJ. TRANCE is necessary and Pols HAP, Van Leeuwen JPTM. Differential expression of estrogen
sufficient for osteoblast-mediated activation of bone resorption. J Exp receptors and mRNA during differentiation on human osteoblast
Med 1998;188:9971001. SV-HFO cells. Endocrinology 1997;138:506770.
[7] Fink L, Seeger W, Ermert L, Hanze J, Stahl U, Grimminger F,
[21] Waters KM, Rickard DJ, Riggs BL, Khosla S, Katzenellenbogen JA,
Kummer W, Bohle RM. Real-time quantitative RT-PCR after laser-
Katzenellenbogen BS, Moore J, Spelsberg TC. Estrogen regulation of
assisted cell picking. Nature Med 1998;4:1329 33.
human osteoblast function is determined by the stage of differentia-
[8] Simonet WS, Lacey DL, Dunstan CR, Kelly M, Chang MS, Luthy R,
tion and the estrogen receptor isoform. J Cell Biochem 2001;83:448
Nguyen HQ, Wooden S, Bennett L, Boone T, Shimamoto G, DeRose
62.
M, Elliot R, Colombero A, Tan HL, Trail G, Sullivan J, Davey E,
[22] Bonnleye E, Aubin JE. Differential expression of estrogen receptor-
Bucay N, Renshaw-Gegg L, Hughes TM, Hill D, Pattison W, Camp-
related receptor and estrogen receptor and in osteoblasts in vivo
bell P, Sander S, Van G, Tarpley J, Derby P, Lee R, Boyle WJ, &
Amgen EST Programme. Osteoprotegerin a novel secreted protein and in vitro. J Bone Miner Res 2002;17:13921400.
involved in the regulation of bone density. Cell 1997;89:309 19. [23] Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli C,
[9] Tsuda E, Goto M, Mochizuki S-I, Yano K, Kobayashi F, Morinaga T, Scully S, Tan HL, Xu W, Lacey DL, Boyle WJ, Simonet WS.
Higashio K. Isolation of a novel cytokine from human fibroblasts that Osteoprotegerin-deficient mice develop early onset osteoporosis and
specifically inhibits osteoclastogenesis. Biochem Biophys Res Com- arterial calcification. Genes Dev 1998;12:1260 8.
mun 1997;234:137 42. [24] Coucher PI, Shipman CM, Lippitt J, Perry M, Asosingh K, Hijzen A,
[10] Anderson DM, Maraskovsky E, Billingsley WL, Dougall WC, Tom- Brabbs AC, Van Beek EJR, Holen I, Skerry TM, Dunstan CR, Russell
etsko ME, Roux ER, Teepe MC, DuBose RF, Cosman D, Galibert L. GR, Van Camp B, Vanderkerken K. Osteoprotegerin inhibits the
A homologue of the TNF receptor and its ligand enhance T-cell development of osteolytic bone disease in multiple myeloma. Blood
growth and dendritic-cell function. Nature 1999;390:1759. 2001;98:3534 40.
[11] Bieche I, Parfait B, Laurendeau I, Girault I, Vivaud M, Lidereau. [25] Collin-Osdoby P, Rothe L, Anderson F, Nelson M, Maloney W,
Quantification of estrogen receptor and expression in sporadic Osdoby P. Receptor activator of NF-B and osteoprotegerin expres-
breast cancer. Oncogene 2001;20:8109 55. sion by human microvascular endothelial cells, regulation by inflam-
[12] Hofbauer LC, Dunstan CR, Spelsberg TC, Riggs BL, Khosla S. matory cytokines, and role in human osteoclastogenesis. J Biol Chem
Osteoprotegerin production by human osteoblast lineage cells is stim- 2001;276:20659 72.

Вам также может понравиться