10
PRECLINICAL STUDY RANKL-dependent and RANKL-independent mechanisms of macrophage-osteoclast differentiation in breast cancer Y. S. Lau L. Danks S. G. Sun S. Fox A. Sabokbar A. Harris N. A. Athanasou Received: 16 July 2006 / Accepted: 24 October 2006 / Published online: 7 December 2006 Ó Springer Science+Business Media B.V. 2006 Abstract The cellular and humoral mechanisms accounting for tumour osteolysis in metastatic breast cancer are uncertain. Osteoclasts, the specialised mul- tinucleated cells responsible for tumour osteolysis, are derived from monocyte/macrophage precursors. Breast cancer-derived tumour-associated macrophages (TAMs) are capable of osteoclast differentiation but the cellular and humoral mechanisms controlling this activity are uncertain. In this study, TAMs were iso- lated from primary breast cancers and cultured in the presence and absence of cytokines/growth factors influencing osteoclastogenesis. Extensive TAM-osteo- clast differentiation occurred only in the presence of RANKL and M-CSF; this process was inhibited by OPG and RANK:Fc, decoy receptors for RANKL. Breast cancer-derived fibroblasts and human bone stromal cells expressed mRNA for RANKL, OPG and TRAIL, and co-culture of these fibroblasts with human monocytes stimulated osteoclast formation by a RANKL-dependent mechanism. Osteoclast formation and lacunar resorption also occurred by a RANKL- independent mechanism when the conditioned med- ium from breast cancer cells, MDA-MB-231 and MCF- 7, was added (with M-CSF) to monocyte cultures. Our findings indicate that TAMs in breast cancer are capable of osteoclast differentiation and that breast cancer-derived fibroblasts and breast cancer cells con- tribute to this process by producing soluble factors that influence osteoclast formation by RANKL-dependent and RANKL-independent mechanisms respectively. Keywords Breast cancer Á Osteoclast Á Bone resorption Á RANKL Introduction Skeletal metastasis is a relatively common complica- tion in patients with cancer of the breast. These met- astatic lesions are usually osteolytic and may cause bone pain, pathological fracture and hypercalcaemia [1]. The cellular and molecular mechanisms whereby this tumour osteolysis is effected are uncertain. Breast cancer cells are not capable of lacunar bone resorption and it is thought that tumour osteolysis is effected by stimulating the formation and activity of osteoclasts, multinucleated cells which are specialised to carry out lacunar bone resorption [2, 3]. Osteoclasts are part of the mononuclear phagocyte system and are formed by fusion of mononuclear pre- cursors of haematopoietic origin [4]. In both mouse and man, mononuclear osteoclast precursors circulate in the monocyte fraction and express a monocyte/ macrophage antigenic phenotype [5]. Osteoclast Y. S. Lau Á L. Danks Á A. Sabokbar Á N. A. Athanasou (&) Department of Pathology, Nuffield Department of Orthopaedic Surgery, University of Oxford, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK e-mail: [email protected] S. G. Sun Department of Orthopaedics, Tangdu Hospital, The Fourth Military Medical University, Xian 710038, China S. Fox Department of Cellular Pathology, John Radcliffe Hospital, Oxford OX3 7DU, UK A. Harris Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, UK 123 Breast Cancer Res Treat (2007) 105:7–16 DOI 10.1007/s10549-006-9438-y

RANKL-dependent and RANKL-independent mechanisms of macrophage-osteoclast differentiation in breast cancer

  • Upload
    oxford

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

PRECLINICAL STUDY

RANKL-dependent and RANKL-independent mechanismsof macrophage-osteoclast differentiation in breast cancer

Y. S. Lau Æ L. Danks Æ S. G. Sun Æ S. Fox ÆA. Sabokbar Æ A. Harris Æ N. A. Athanasou

Received: 16 July 2006 / Accepted: 24 October 2006 / Published online: 7 December 2006� Springer Science+Business Media B.V. 2006

Abstract The cellular and humoral mechanisms

accounting for tumour osteolysis in metastatic breast

cancer are uncertain. Osteoclasts, the specialised mul-

tinucleated cells responsible for tumour osteolysis, are

derived from monocyte/macrophage precursors. Breast

cancer-derived tumour-associated macrophages

(TAMs) are capable of osteoclast differentiation but

the cellular and humoral mechanisms controlling this

activity are uncertain. In this study, TAMs were iso-

lated from primary breast cancers and cultured in the

presence and absence of cytokines/growth factors

influencing osteoclastogenesis. Extensive TAM-osteo-

clast differentiation occurred only in the presence of

RANKL and M-CSF; this process was inhibited by

OPG and RANK:Fc, decoy receptors for RANKL.

Breast cancer-derived fibroblasts and human bone

stromal cells expressed mRNA for RANKL, OPG and

TRAIL, and co-culture of these fibroblasts with human

monocytes stimulated osteoclast formation by a

RANKL-dependent mechanism. Osteoclast formation

and lacunar resorption also occurred by a RANKL-

independent mechanism when the conditioned med-

ium from breast cancer cells, MDA-MB-231 and MCF-

7, was added (with M-CSF) to monocyte cultures. Our

findings indicate that TAMs in breast cancer are

capable of osteoclast differentiation and that breast

cancer-derived fibroblasts and breast cancer cells con-

tribute to this process by producing soluble factors that

influence osteoclast formation by RANKL-dependent

and RANKL-independent mechanisms respectively.

Keywords Breast cancer � Osteoclast � Bone

resorption � RANKL

Introduction

Skeletal metastasis is a relatively common complica-

tion in patients with cancer of the breast. These met-

astatic lesions are usually osteolytic and may cause

bone pain, pathological fracture and hypercalcaemia

[1]. The cellular and molecular mechanisms whereby

this tumour osteolysis is effected are uncertain. Breast

cancer cells are not capable of lacunar bone resorption

and it is thought that tumour osteolysis is effected by

stimulating the formation and activity of osteoclasts,

multinucleated cells which are specialised to carry out

lacunar bone resorption [2, 3].

Osteoclasts are part of the mononuclear phagocyte

system and are formed by fusion of mononuclear pre-

cursors of haematopoietic origin [4]. In both mouse

and man, mononuclear osteoclast precursors circulate

in the monocyte fraction and express a monocyte/

macrophage antigenic phenotype [5]. Osteoclast

Y. S. Lau � L. Danks � A. Sabokbar � N. A. Athanasou (&)Department of Pathology, Nuffield Department ofOrthopaedic Surgery, University of Oxford, NuffieldOrthopaedic Centre, Oxford OX3 7LD, UKe-mail: [email protected]

S. G. SunDepartment of Orthopaedics, Tangdu Hospital, The FourthMilitary Medical University, Xian 710038, China

S. FoxDepartment of Cellular Pathology, John Radcliffe Hospital,Oxford OX3 7DU, UK

A. HarrisWeatherall Institute of Molecular Medicine, John RadcliffeHospital, Oxford OX3 9DS, UK

123

Breast Cancer Res Treat (2007) 105:7–16

DOI 10.1007/s10549-006-9438-y

differentiation from these mononuclear precursors re-

quires the presence of macrophage-colony stimulating

factor (M-CSF) and involves a receptor-ligand inter-

action with cells of the osteoblast lineage, which ex-

press a membrane-bound osteoclast differentiation

factor termed receptor activator for nuclear factor jB

ligand (RANKL) [6]. RANKL interacts with its

receptor, RANK, which is expressed by mononuclear

osteoclast precursors; this process is inhibited by os-

teoprotegerin (OPG), which is produced by bone

stromal cells and breast cancer cells [7–9]. In addition

to this RANKL-dependent mechanism of osteoclast

formation, it has been shown several cytokines, such as

tumour necrosis factor-a (TNF-a) and interleukin-6

(IL-6), and growth factors, such as transforming growth

factor-b (TGF-b), can induce osteoclast formation

from marrow and circulating osteoclast precursors by a

mechanism independent of RANKL [10–13].

A prominent macrophage infiltrate is commonly

found in both primary and secondary breast cancers

[14, 15]. We have previously shown that TAMs isolated

from primary human and mouse mammary carcinomas

are capable of osteoclast differentiation when these

cells are co-cultured with bone-derived stromal cells in

the presence of 1,25 dihydroxyvitamin D3 and M-CSF

[16, 17]. We have also shown that breast cancer cells

secrete factors that dose-dependently influence human

osteoclast formation [18]. The precise cellular and

molecular mechanisms whereby TAMs in breast can-

cer differentiate into osteoclasts are not known. In this

study, we have analysed the role of RANKL-depen-

dent and RANKL-independent mechanisms in TAM-

osteoclast differentiation in breast cancer. We have

also examined whether the other major cellular com-

ponents found in a breast cancer metastasis (i.e. breast

cancer cells, tumour fibroblasts and bone stromal cells)

influence osteoclast formation from TAMs. As in

previous studies, we isolated TAMs and tumour fi-

broblasts from primary breast cancers rather than

skeletal metastases of breast cancer as the latter would

contain mature bone-resorbing osteoclasts and thus

make it impossible to assess osteoclast formation in

culture.

Materials and methods

This study was approved by the Oxford Clinical Re-

search Ethics Committee. Alpha minimum essential

medium (MEM) and fetal bovine serum (FBS) were

purchased from Gibco Laboratories (Paisley, UK);

MEM containing 10% FBS, 100 U/ml penicillin, and

10 lg/ml streptomycin (MEM/FBS) was used for all

cell culture experiments unless otherwise specified.

Recombinant human M-CSF, OPG, RANK:Fc, and

anti-human TNF-a antibody were obtained from R&D

Systems Europe (Abingdon, UK). Soluble RANKL

was obtained from Peprotech (London, UK). All re-

agents used in reverse transcription and DNA ampli-

fication were obtained from Invitrogen (Paisley, UK).

All cultures were incubated at 37�C in a humidified

atmosphere of 5% CO2 and 95% air, and carried out in

triplicate.

TAMs (and tumour fibroblasts) were isolated from

primary invasive ductal breast carcinomas were ob-

tained from eight female patients (age range 51–76).

TAM isolation and culture

The tumour tissue was washed in sterile phosphate

buffered saline. Fragments of the tumour were then

placed in 1 mg/ml of collagenase Type 1 (Sigma-Al-

drich, Dorset, UK) and incubated for 1 h. The digested

tissue suspension was passed through a Falcon� 70 lm

pore size cell strainer (Becton Dickinson, Oxford,

UK). The filtrate was centrifuged at 1800g for 10 min

and the cell pellet resuspended in 2 ml of MEM/FBS.

The cell yield was determined using a haemocytometer

after lysis of red blood cells with 5% (v/v) acetic acid.

1 · 105 cells per well were added to 96-well tissue

culture plates containing glass coverslips and dentine

slices prepared as previously described [19]. After 2 h

incubation, dentine slices and coverslips were removed

from the wells, washed vigorously in MEM/FBS to

remove non-adherent cells and then placed in a 24-well

tissue culture plate containing 1 ml of MEM/FBS

supplemented with M-CSF (25 ng/ml) and/or RANKL

(30 ng/ml). Negative controls contained no added fac-

tors. All cultures were maintained for 24 h and up to

21 days. Culture medium containing these factors was

replenished every 3–4 days. To determine macrophage

purity in the isolated TAM cell population, 24-h cell

cultures were stained immunohistochemically by an

indirect immunoperoxidase technique with monoclonal

antibody GSR1 (Dakopatts, Glostrup, Denmark) di-

rected against CD14 (a monocyte/macrophage marker)

[20], breast cancer cell markers E29 and MNF116

(Dakopatts, Glostrup, Denmark), directed against

epithelial membrane antigen (EMA) and cytokeratin

respectively.

Isolation and culture of human peripheral blood

mononuclear cells (PBMCs)

Human PBMCs were obtained by density gradient

centrifugation of 50 ml of buffy coat cell preparation

8 Breast Cancer Res Treat (2007) 105:7–16

123

provided by the National Blood Transfusion Service

(Bristol, UK). The buffy coat preparation was mixed

with an equal volume of MEM and purified over

Histopaque (Sigma-Aldrich, Dorset, UK). After cen-

trifugation at 2250 rpm for 25 min, the cell layer above

the Histopaque was collected, suspended in MEM, and

centrifuged at 1800 rpm for 10 min. The cell pellet was

resuspended in MEM and centrifuged again. 5 ml of

MEM/FBS was then added and the number of cells

counted in a haematocytometer following lysis of red

blood cells with 5% (v/v) acetic acid. 5 · 105 cells per

well in 100 ll of MEM/FBS were plated immediately

onto dentine slices and glass coverslips in a 96-well

tissue culture plate. After 3 h incubation, the dentine

slices and glass coverslips were washed in MEM/FBS

to remove any non-adherent cells, and then transferred

to 24-well tissue culture plates containing MEM/FBS

and M-CSF (25 ng/ml). Positive controls were set up in

the presence of M-CSF (25 ng/ml) and RANKL

(30 ng/ml).

Cytochemical and functional assessment

of osteoclast differentiation

Histochemistry for the expression of the osteoclast-

associated enzyme, tartrate-resistant acid phosphatase

(TRAP) was carried out on 14-day cell cultures on

glass coverslips using a commercially available kit

(Sigma-Aldrich, Dorset, UK) [21]. These cell cultures

were also stained immunohistochemically with mono-

clonal antibody 23C6 (Serotec, Oxford, UK) directed

against the vitronectin receptor (VNR) (an osteoclast-

associated antigen) [22].

Functional evidence of osteoclast differentiation was

determined by a lacunar resorption assay system using

cell cultures on dentine slices as previously described

[19]. After 21-day incubation, the cells were removed

from the dentine slices by treatment with 1 M ammo-

nium hydroxide. The dentine slices were washed in

distilled water, ultrasonicated to remove adherent

cells, then stained with 0.5% (w/v) toluidine blue to

reveal areas of lacunar resorption and examined by

light microscopy.

Generation of breast cancer-derived fibroblasts

and human bone stromal cells

Following collagenase digestion of the tumour tissue,

isolated cells were suspended in MEM/FBS and placed

in 25 cm2 tissue culture flasks and incubated for up to

3 weeks. The medium was changed after 24-h incuba-

tion and then at 5–7 day intervals until the cell

cultures were confluent. These cultures, containing

spindle-shaped fibroblast-like cells, were passaged by

treatment with trypsin (0.25%)/EDTA (1 mM) at least

3 times before removal in preparation for RNA

extraction and co-culture experiments.

Cultures of bone stromal cells were also derived

from explants of femoral cancellous bone derived from

patients undergoing hip arthroplasty for osteoarthritis

as previously described [23]. The bone pieces were cut

into small fragments, washed vigorously in sterile PBS

to remove blood and fat, then suspended in MEM/FBS

and placed in 25 cm2 tissue culture flasks. The medium

was changed after 24-h incubation and subsequently at

5–7 day intervals. These cultures, containing spindle-

shaped cells, were passaged twice before being re-

moved and used for RNA extraction.

Both tumour-derived fibroblasts and bone stromal

cell cultures were stained for alkaline phosphatase, an

osteoblast-associated marker, and immunohistochemi-

cally with antibodies directed against prolyl-4-hydrox-

ylase and vimentin (both from Dakopatts); these

antigenic markers are expressed by both fibroblasts

and osteoblasts. The fibroblast and osteoblast cultures

were also stained immunohistochemically for leucocyte

common antigen, using monoclonal antibodies PD7/26

(Dakopatts), as well as for TRAP, CD14 and VNR as

described above.

Breast cancer-derived fibroblast total RNA

extraction and RT-PCR

Total RNA extraction was carried out using the

RNeasy� mini kit (QIAGEN, Hombrechtikon, Swit-

zerland), according to the manufacturer’s instructions.

Single strand complementary DNA (cDNA) was syn-

thesised from 2.0 lg of total RNA according to stan-

dard protocols using the SuperScript� First-Strand

Synthesis System for RT-PCR. cDNA was amplified by

PCR to generate products corresponding to messenger

RNA (mRNA) encoding human gene products for

GAPDH, RANKL, OPG and TRAIL (Table 1).

Aliquots of PCR products were fractionated on 1%

agarose gels stained with ethidium bromide. Gel pic-

tures and quantification of signals were obtained after

scanning with AlphaImager 2200 (Alpha Innotech

Corporation, USA) and ImageJ software analysis

(public domain Java image processing program).

Co-culture of PBMCs and breast cancer-derived

fibroblasts/bone stromal cells

Breast cancer-derived fibroblasts, harvested as previ-

ously described, were seeded at 1 · 104 cells per well

Breast Cancer Res Treat (2007) 105:7–16 9

123

onto PBMCs prepared as described above, and sup-

plemented with the following factors:

(1) M-CSF (25 ng/ml)

(2) M-CSF (25 ng/ml) and OPG (500 ng/ml)

(3) M-CSF (25 ng/ml) and RANK:Fc (500 ng/ml)

Parallel co-culture experiments were set up with

human bone stromal cells. All cultures were main-

tained for 24 h, 14 and 21 days. Culture medium and

factors was replenished every 3–4 days.

Effect of breast cancer cells on osteoclast formation

Breast cancer cell conditioned medium (CM) was ob-

tained from cultures of the human breast cancer cell

lines, MDA-MB-231 and MCF-7. This was added to

human PBMCs plated onto glass coverslips and den-

tine slices, prepared as described above, in a 24-well

tissue culture plate containing 1 ml of MEM/FBS,

subjected to one of the following treatments:

(a) 0–50% breast cancer cell CM and M-CSF (25 ng/

ml)

(b) 0–50% breast cancer cell CM, M-CSF (25 ng/ml)

and RANKL (30 ng/ml)

(c) 10% breast cancer cell CM, M-CSF (25 ng/ml)

and anti-human TNF-a antibody (10 lg/ml)

(d) 10% breast cancer cell CM, M-CSF (25 ng/ml)

and RANK:Fc (500 ng/ml)

Cultures on coverslips and dentine slices were

maintained for 14 and 21 days respectively, with cul-

ture medium, CM and factors replenished every 3–

4 days.

Statistical analysis

The extent of lacunar resorption was measured using

an image analysis software (Adobe Photoshop, USA)

as previously [17] described, and expressed as the mean

percentage of surface area resorbed (%SA) ± standard

error of mean (SEM). In order to minimise the effect

of batch-to-batch variation of PBMCs, all resorption

data were normalised and expressed relative to the

response obtained in PBMC cultures incubated with

25 ng/ml M-CSF and 30 ng/ml RANKL (positive con-

trol). Statistical significance was determined using the

unpaired t-test and P values <0.05 were considered

significant.

Results

Characterisation of TAMs isolated from breast

cancer

After 24-h incubation in the presence or absence of

RANKL and M-CSF, TAMs isolated from all eight tu-

mours strongly expressed CD14 (Fig. 1), a macrophage

antigen which is known not to be present on osteoclasts.

These cells were negative for the osteoclast markers,

TRAP and VNR and the breast cancer cell markers,

EMA and cytokeratin. 24-h TAM cultures on dentine

slices, both in the presence and absence of M-CSF and

Table 1 Human primer sequences used in amplification

Primer sequence Size of product(base pairs)

Annealingtemp. (�C)

GAPDH forward 5¢-CAC TGA CAC GTT GGC AGT GG-3¢reverse 5¢-CAT GGA GAA GGC TGG GGC TC-3¢

360 60

OPG forward 5¢- ATG AAC AAG TTG CTG TGC TG-3¢reverse 5¢-GCA GAA CTC TAT CTC AAG GTA-3¢

354 58

RANKL forward 5¢-CAG ATG GAT CCT AAT AGA AT-3¢reverse 5¢-ATG GGA ACC AGA TGG GAT GTC-3¢

324 56

TRAIL forward 5¢-ATC ATG GCT ATG ATG GAG GT-3¢reverse 5¢-AAC TGT AGA AAT GGT TTC CTC-3¢

315 58

Fig. 1 Day 1 TAMs isolated from melanoma strongly expressmacrophage marker CD14. Bar = 50 lm

10 Breast Cancer Res Treat (2007) 105:7–16

123

RANKL, also showed no evidence of lacunar resorp-

tion. The mononuclear cells isolated from these

tumours thus only expressed the phenotypic markers of

macrophages and not osteoclasts or tumour cells.

TAM-osteoclast differentiation is mediated through

RANKL

In the presence of RANKL and M-CSF, numerous

multinucleated cells, expressing the osteoclast-associ-

ated markers, TRAP and VNR, were formed in 14-day

TAM cultures incubated on glass coverslips (Fig. 2A,

B) in all eight cases. No expression of TRAP or VNR

was seen when either RANKL or M-CSF was omitted.

Scattered mononuclear and multinucleated cells posi-

tive for CD14 were also noted in these cultures, indi-

cating that not all TAMs incubated with RANKL and

M-CSF underwent osteoclast differentiation.

In 21-day TAM cultures on dentine slices incubated

with M-CSF and RANKL, functional evidence of

osteoclast differentiation was noted with the formation

of numerous areas of lacunar resorption in the all

cases; these were largely in the form of multiple com-

pound areas of lacunar excavation on the dentine

surface (Fig. 2C). In the absence of either M-CSF or

soluble RANKL, lacunar resorption was not seen.

Characterisation of breast cancer-derived

fibroblasts and bone stromal cells

After 3–4 passages, the cells which were isolated from

breast cancers consisted almost entirely of spindle-

shaped, fibroblast-like mononuclear cells. These cells

did not stain positively for EMA, cytokeratin, CD45,

CD14, VNR or TRAP, indicating that these cultures

did not contain tumour cells, macrophages or osteo-

clasts; cultured cells were positive for vimentin and

prolyl-4-hydroxylase but negative for alkaline phos-

phatase (Fig. 3A, B). Bone stromal cells cultured from

femoral bone explants showed similar morphological

characteristics and were positive for alkaline

phosphatase. Multinucleated cells did not form in

fibroblast or bone stromal cell cultures and no

resorption was seen on dentine slices on which only

fibroblasts or bone marrow stromal cells were cultured.

Using a semi-quantitative RT-PCR method, signals

generated by mRNA levels of RANKL, OPG and

TRAIL were quantified relative to GAPDH. This

showed that mRNA for RANKL, OPG and TRAIL

was expressed by bone stromal cells and cultured

breast cancer-derived fibroblasts in all cases studied

(Fig. 3C).

Effect of breast cancer-derived fibroblasts

and breast cancer cells on osteoclast formation

Co-cultures of breast cancer-derived fibroblasts and

human monocytes in the presence of M-CSF resulted

in the formation of TRAP+ and VNR+ multinucleated

cells capable of lacunar resorption (Fig. 4). The addi-

tion of OPG or RANK:Fc to these co-cultures abol-

ished osteoclast formation and resorption. As

previously shown [24], co-cultures of human bone

stromal cells and monocytes in the presence of M-CSF,

also resulted in the formation of TRAP+/VNR+ mul-

tinucleated cells capable of lacunar resorption. The

addition of CM from cultured breast cancer-derived

fibroblasts to monocytes did not induce osteoclast

formation.

Cultures of human PBMCs incubated with M-CSF

and CM from the breast cancer cell lines MCF-7 and

MDA-MB-231 resulted in the generation of mononu-

clear and small multinucleated (<4 nuclei) TRAP+ and

VNR+ cells (Fig. 5A, B) capable of forming a few

small round or ovoid lacunar resorption pits (Fig. 5C);

large areas of compound lacunar excavation, as noted

in M-CSF and RANKL-treated positive controls, were

not seen (Fig. 5D). The formation of these resorption

pits was not abolished by the addition of OPG,

RANK:Fc or a neutralising antibody to TNF-a. The

addition of RANKL to human PBMC cultures incu-

bated with M-CSF and CM for breast cancer cell lines

Fig. 2 (A) TRAP positive and (B) VNR positive multinucleatedcells in 14-day TAM cultures in the presence of M-CSF andRANKL and (C) compound areas of lacunar resorption on

dentine slices in 21-day TAM cultures under similar conditions(Toluidine blue staining) (Bars = 50 lm)

Breast Cancer Res Treat (2007) 105:7–16 11

123

MCF-7 and MDA-MB-231 showed a dose-dependent

inhibition of osteoclast formation and lacunar resorp-

tion with formation of fewer TRAP+ multinucleated

cells and fewer resorption pits in 14- and 21-day

cultures respectively (relative to positive control)

(Fig. 6).

Fig. 3 (A) Fibroblastsderived form breast cancerstaining positive for vimentinand negative for cytokeratin.Bar = 50 lm. (B) Expressionof RANKL, OPG andTRAIL mRNA by fibroblastsderived from breast cancer.Reverse transcription-polymerase chain reactionproducts were fractionated onagarose gel. Lane 1, positivecontrol (+ctl); lane 2, negativecontrol (–ctl); lanes 3–8,breast cancer fibroblasts from6 patients (F1–6); lanes 9–11,normal bone marrow stromalcells from 3 patients (N1–3)

Fig. 4 (A) TRAP positivemultinucleated cell and (B)lacunar resorption on adentine slice (Toluidine bluestaining) in co-cultures ofbreast cancer-derivedfibroblasts and humanmonocytes incubated in thepresence of M-CSF(Bars = 25 lm)

Fig. 5 (A) TRAP and (B)VNR positive cells in 14-dayhuman monocyte culture inthe presence of M-CSF andMCF-7 conditioned medium.(C) Few small, round or ovoidlacunar resorption pits ondentine slices formed in 21-day human monocyte culturein the presence of M-CSF andMCF-7 conditioned medium,were unlike (D) large,compound lacunar excavationseen in RANKL-treatedmonocyte cultures [Toluidineblue staining (A) and (B)](Bars = 100 lm)

12 Breast Cancer Res Treat (2007) 105:7–16

123

Discussion

Bone metastases in breast cancer are commonly

osteolytic, being associated with marked osteoclastic

bone resorption [3]. These metastatic deposits contain

numerous macrophages (i.e. TAMs) as well as other

cellular components including tumour cells and fibro-

blasts in the tumour stroma. In this study, we have

shown that one means whereby tumour osteolysis is

effected in a breast cancer metastasis to bone is by

TAM-osteoclast differentiation. We found that large

numbers of osteoclasts and numerous lacunar resorp-

tion pits were formed when breast cancer-derived

TAMs were cultured with RANKL and M-CSF. We

also noted that fibroblasts derived from breast cancers,

like human bone stromal cells, expressed RANKL and

were capable of supporting monocyte-osteoclast dif-

ferentiation. These findings suggest that RANKL-in-

duced TAM-osteoclast differentiation most likely

involves an interaction with these cellular components

of a skeletal metastasis. Breast cancer cells did not

stimulate RANKL-induced osteoclast formation but

were found to produce a soluble factor that could in-

duce the formation of osteoclasts from mononuclear

phagocytes by a RANKL-independent mechanism.

Tumor-associated macrophages are a major com-

ponent of the inflammatory cell infiltrate within and

around primary and metastatic tumours [14, 15]. A

relatively high macrophage: tumour cell ratio is seen at

sites of skeletal metastases where osteolysis is occur-

ring rapidly [25–27]. Tumour cells are known to secrete

several factors which induce macrophage recruitment

into tumours, including monocyte chemotactic protein-

1 and M-CSF [28–30]. Proliferation and survival of

TAMs has been related to tumour cell production of

M-CSF, and TAMs are known to express the c-fms

proto-oncogene, which encodes the M-CSF receptor

[28, 31]. Previous studies have shown that murine and

human TAMs, isolated from primary breast carcino-

mas, are capable of differentiation into TRAP+

osteoclastic cells capable of extensive lacunar resorp-

tion [16, 17, 19]. Osteoclast differentiation involves an

interaction between RANK-expressing cells of the

monocyte-macrophage lineage and RANKL-express-

ing bone stromal cells, a process that is inhibited by

OPG [6–8]. In this study, we have shown that TAMs

isolated from primary breast carcinomas are capable of

osteoclast differentiation in the presence of RANKL

and M-CSF. TAMs isolated from carcinomas ex-

pressed the monocyte/macrophage marker CD14 and

were negative for the osteoclast markers TRAP and

VNR; these cells, like human monocytes, differentiate

into TRAP+ and VNR+ multinucleated cells capable

of carrying out lacunar resorption when incubated with

RANKL and M-CSF [32–34]. This is likely to be the

principal means whereby osteoclasts are formed from

TAMs in a skeletal metastasis as the lacunar resorption

seen in RANKL-treated monocyte cultures was

extensive and characterised by the formation of

numerous compound lacunar resorption pits.

A breast cancer metastasis in bone contains not only

tumour cells and TAMs but also connective tissue cells,

including fibroblasts in the tumour stroma and bone

stromal cells/osteoblasts in the bone itself. Bone stro-

mal cells and osteoblasts, as well as some fibroblast

populations, are known to express RANKL and OPG

[35–37]. In this study we have shown that fibroblasts

derived from primary breast carcinomas express

RANKL, OPG and TRAIL; the latter is known to bind

OPG. We found that breast cancer-derived fibroblasts,

like bone stromal cells, supported monocyte-osteoclast

differentiation; this process was abolished by the

addition of OPG and RANK:Fc, indicating that these

stromal cells induced osteoclast formation by a

Fig. 6 % Surface area (SA) resorption formed in human PBMCcultures incubated with M-CSF, RANKL and breast cancer cellline CM relative to positive control (PBMC cultures with M-CSFand RANKL). Error bars denote SEM (n = 5). [*(P < 0.05),**(P < 0.005) and ***(P < 0.0001) denote significant differencein lacunar resorption relative to positive control]

Breast Cancer Res Treat (2007) 105:7–16 13

123

RANKL-dependent mechanism. Differential expres-

sion and release of RANKL, OPG and TRAIL by

tumour fibroblasts and bone stromal cells may play a

role in determining the extent of osteoclast formation

and osteolysis that occurs in metastatic carcinomas.

OPG is expressed at high concentration in a wide range

of tissues [7]. OPG has broad binding specificity and

binds to TRAIL [38]. RANKL is known to exist in cell

membrane-bound and soluble forms but is mainly a

membrane-bound protein in vivo [7]. In contrast,

RANKL is most abundant in skeletal and lymphoid

tissues. It is likely that the RANKL:OPG ratio in dif-

ferent tissues determines the extent of osteoclast for-

mation and resorption activity.

The other major cellular component of a metastatic

breast carcinoma in bone is, of course, the breast

cancer cells themselves. The expression of osteoclas-

togenic factors in breast cancers is controversial. Tho-

mas et al found that breast cancer cells expressed OPG

and TRAIL but not RANKL, and that they did not

support osteoclast formation in co-culture with marrow

haematopoietic precursors [9]. In contrast, others have

found expression of RANKL and TRAIL in some

breast cancers [39]. If this is the case, then TRAIL may

modulate RANKL-induced osteoclastogenesis. Breast

cancer cells are also known to produce numerous

cytokines and growth factors, such as IL-1, IL-6, TNF-aand PTHrP, which promote RANKL but inhibit OPG

expression by cells of the osteoblast lineage [7]. In this

study, we found that the CM derived from cultured

MCF-7 and MDA-MB-231 breast cancer cells did not

stimulate (and actually inhibited) osteoclast formation

when added to RANKL-treated monocyte cultures,

but that this CM could induce osteoclast formation in

the absence of RANKL. CM-induced inhibition of

RANKL-induced osteoclastogenesis may have been

due to the presence of OPG in the CM; breast cancer

cells are known to produce OPG [9], and this would

effectively have neutralised the osteoclastogenic effect

of the soluble RANKL added to monocyte cultures.

The addition of breast cancer CM alone (i.e. in the

absence of RANKL), however, to monocyte cultures

resulted in the formation of small TRAP+ and VNR+

osteoclastic cells that were capable of lacunar resorp-

tion. This process was not inhibited by OPG or

RANK:Fc, indicating that the soluble osteoclastogenic

factor produced by cultured breast cancer cells acted

by a RANKL-independent mechanism.

In contrast to the numerous large osteoclasts and

extensive areas of lacunar excavation produced when

monocytes and TAMs were cultured with RANKL, the

osteoclasts formed in cultures to which breast cancer

CM was added were small; these cells contained fewer

than 4 nuclei and were associated with the formation of

relatively small areas of lacunar resorption. TNF-a and

other growth factors/cytokines known to induce

RANKL-independent osteoclastogenesis typically

generate small TRAP+ and VNR+ osteoclasts that

form relatively small resorption pits [10–13]. We found

that the addition of a neutralising antibody to TNF-adid not abolish osteoclast formation or lacunar

resorption pit formation in monocyte cultures con-

taining breast cancer CM. As breast cancer cells are

known to produce a large number of cytokines and

growth factors [40], it is possible that one or more of

the humoral factors known to stimulate RANKL-

independent osteoclast formation in this way remained

operative, resulting in the formation of mature func-

tional osteoclasts. We have recently reported that

melanoma cells also secrete a soluble factor (>10 kDa)

which promotes osteoclast formation by a RANKL-

independent mechanism [41].

In summary, we have shown that the cellular com-

ponents of a metastatic breast carcinoma play a role in

promoting the osteoclast formation that is required for

tumour osteolysis. Osteoclasts are formed from TAMs

in breast cancer and this can occur by both RANKL-

dependent and RANKL-independent mechanisms; the

former involves an interaction between RANK-

expressing mononuclear phagocyte osteoclast precur-

sors, which are present in the TAM population, and

RANKL-expressing host bone stromal cells and tu-

mour fibroblasts. Osteoclast formation can also occur

by a RANKL-independent mechanism through breast

cancer cell secretion of one or more soluble osteo-

clastogenic factors. The contribution of RANKL-

dependent and RANKL-independent mechanisms of

pathological bone resorption will need to be taken into

account in devising therapies to treat tumour osteolysis

due to skeletal breast cancer metastasis.

Acknowledgements The authors wish to thank the Frances andAugustus Newman Foundation, Jenny Mays-Smith Skin CancerResearch Fund, Oxfordshire Health Service Research Commit-tee and the Rosetrees Charitable Trust.

References

1. Stoll BA (1983) Natural history, prognosis and staging ofbone metastases. In: Stoll BA, Parbhoo S (eds) Bonemetastases: monitoring and treatment. Raven Press, NewYork, pp 1–4

2. Clohisy DR, Palkert D, Ramnaraine ML, Pekurovsky I,Oursler MJ (1996) Human breast cancer induces osteoclastactivation and increases the number of osteoclasts at sites oftumor osteolysis. J Orthop Res 14:396–402

3. Mundy GR (1991) Mechanisms of osteolytic bone destruc-tion. Bone 12:51–56

14 Breast Cancer Res Treat (2007) 105:7–16

123

4. Blair HC, Athanasou NA (2004) Recent advances in osteo-clast biology. Histol and Histopathol 19:189–199

5. Fujikawa Y, Quinn JM, Sabokbar A, McGee JO, AthanasouNA (1996) The human osteoclast precursor circulates in themonocyte fraction. Endocrinology 137:4058–4060

6. Yasuda H, Shima N, Nakagawa N, Yamaguchi K, KinosakiM, Mochizuki S, Tomoyasu A, Yano K, Goto M, MurakamiA, Tsuda E, Morinaga T, Higashio K, Udagawa N, Takah-ashi N, Suda T (1998) Osteoclast differentiation factor is aligand for osteoprotegerin/osteoclast inhibitory factor and isidentical to TRANCE/RANKL. Proc Natl Acad Sci USA95:3597–3602

7. Hofbauer LC, Khosla S, Dunstan C, Lacey DL, Boyle W,Riggs BL (2000) The roles of osteoprotegerin and osteo-protegerin ligand in the paracrine regulation of boneresorption. Bone Min Res 15:2–12

8. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS,Luthy R, Nguyen HQ, Wooden S, Bennett L, Boone T,Shimamoto G, DeRose M, Elliott R, Colombero A, Tan HL,Trail G, Sullivan J, Davy E, Bucay N, Renshaw-Gegg L,Hughes TM, Hill D, Pattison W, Campbell P, Boyle WJ, et al(1997) Osteoprotegerin: a novel secreted protein involved inthe regulation of bone density. Cell 89:309–319

9. Thomas RJ, Guise TA, Yin JJ, Elliott J, Horwood NJ,Martin TJ, Gillespie MT (1999) Breast cancer cells interactwith osteoblasts to support osteoclast formation. Endocri-nology 140:4451–4458

10. Itonaga I, Kudo O, Sabokbar A, Danks L, Ferguson D,Fujikawa Y, Athanasou NA (2004) Transforming growthfactor b induces human osteoclast formation. Bone 34:57–64

11. Kobayashi K, Takahashi N, Jimi E, Udagawa N, Takami M,Kotake S, Nakagawa N, Kinosaki M, Yamaguchi K, ShimaN, Yasuda H, Morinaga T, Higashio K, Martin TJ, Suda T(2000) Tumor necrosis factor alpha stimulates osteoclastdifferentiation by a mechanism independent of the ODF/RANKL-RANK interaction. J Exp Med 191:275–286

12. Kudo O, Fujikawa Y, Itonaga I, Sabokbar A, Torisu T, A-thanasou NA (2002) Proinflammatory cytokine (TNFalpha/IL-1alpha) induction of human osteoclast formation. J Pa-thol 198:220–227

13. Kudo O, Sabokbar A, Pocock A, Itonaga I, Fujikawa Y,Athanasou NA (2003) Interleukin-6 and interleukin-11 sup-port human osteoclast formation by a RANKL-independentmechanism. Bone 32:1–7

14. Bugelski PJ, Corwin SP, North SM, Kirsh RL, NicolsonGL, Poste G (1987) Macrophage content of spontaneousmetastases at different stages of growth. Cancer Res47:4141–4145

15. Van Ravenswaay Classen HH, Kluin PM, Fleuren G (1992)Tumour infiltrating cells in human cancer: on the possiblerole of CD16+ macrophages in anti-tumour cytoxicity. LabInvest 67:166–174

16. Quinn JM, Athanasou NA, McGee JO’D (1992) Tumourinfiltrating macrophages are capable of bone resorption. JCell Science 101:681–686

17. Quinn J, Matsumura Y, Tarin D, McGee JO’D, AthanasouNA (1994) Cellular and hormonal mechanisms associatedwith malignant bone resorption. Lab Invest 71:465–471

18. Hunt N, Fujikawa Y, Itonaga I, Harris A, Athanasou NA(2001) Cellular mechanisms of bone resorption in breastcancer. Br J Cancer 85:78–84

19. Quinn JMW, McGee JO’D, Athanasou NA (1998) Humantumour-associated macrophages differentiate into osteo-clastic bone-resorbing cells. J Pathol 184:31–36

20. Athanasou NA, Quinn J (1990) Immunophenotypic differ-ences between osteoclasts and macrophage polykaryons:

immunohistological distinction and implications for osteo-clast ontogeny and function. J Clin Pathol 43:997–1003

21. Minkin C (1982) Bone acid phosphatase: tartrate-resistantacid phosphatase as a marker of osteoclast function. CalcifTissue Int 34:285–290

22. Davies J, Warwick J, Totty N, Philp R, Helfrich M, HortonM (1989) The osteoclast functional antigen, implicated in theregulation of bone resorption, is biochemically related to thevitronectin receptor. J Cell Biol 109:1817–1826

23. Gundle R, Beresford JN (1995) The isolation and culture ofcells from explants of human trabecular bone. Calcif TissueInt 56:8–10

24. Sun SG, Lau YS, Itonaga I, Sabokbar A, Athanasou NA(2006) Bone stromal cells in pagetic bone and Paget’s sar-coma express RANKL and support human osteoclast for-mation. J Pathol 209(1):114–120

25. Bugelski PJ, Kirsh RL, Sowinski JM, Poste GL (1985)Changes in the macrophage content of lung metastases atdifferent stages in tumor growth. Am J Pathol 118:419–424

26. Normann SJ (1985) Macrophage infiltration and tumourprogression. Cancer Metastasis Rev 4:227–291

27. Galasko CBS (1976) Mechanisms of bone destruction in thedevelopment of skeletal metastasis. Nature 276:726–728

28. Mantovani A (1994) Tumor-associated macrophages inneoplastic progression: a paradigm for the in vivo function ofchemokines. Lab Invest 71:5–16

29. Yamashiro S, Takaya M, Nishi T, Kuratsu J, Yashimura T,Usho Y, Takahashi K (1994) Tumor-derived monocytechemoattractant protein induces intratumoral infiltration ofmonocyte-derived macrophage subpopulation in trans-planted rat tumour. Am J Pathol 145:856–867

30. Yoshimura T, Takeya M, Takahashi K (1991) Molecularcloning of rat monocyte chemoattractant protein-1 (MCP-1)and its expression in rat spleen cells and tumour cell lines.Biochem Biophys Res Commun 174:504–509

31. Flanagan AM, Lader CS (1998) Update on the biologic ef-fects of macrophage-colony stimulating factor. Curr OpinionHematol 5:181–185

32. Faust J, Lacey DL, Hunt P, Burgess TL, Scully S, Van G, EliA, Qian Y, Shalhoub V (1999) Osteoclast markers accumu-late on cells developing from human peripheral bloodmononuclear precursors. J Cell Biochem 72:67–80

33. Quinn JWM, Elliott J, Gillespie MT, Martin TJ (1998) Acombination of osteoclast differentiation factor and macro-phage-colony stimulating factor is sufficient for both humanand mouse osteoclast formation. Endocrinology 139:4424–4427

34. Massey HM, Flanagan AM (1999) Human osteoclasts derivefrom CD14-positive monocytes. Br J Haematol 106:167–170

35. Quinn JM, Horwood NJ, Elliott J, Gillespie MT, Martin TJ(2000) Fibroblastic stromal cells express receptor activator ofNF-kappa B ligand and support osteoclast differentiation. JBone Miner Res 15:1459–1466

36. Sabokbar A, Itonaga I, Sun SG, Kudo O, Athanasou NA(2005) Arthroplasty membrane-derived fibroblasts directlyinduce osteoclast formation and osteolysis in aseptic loos-ening. J Orthop Res 23:511–519

37. Takayanagi H, Iizuka H, Juji T, Nakagawa T, Yamamoto A,Miyazaki T, Koshihara Y, Oda H, Nakamura K, Tanaka S(2000) Involvement of receptor activator of nuclear factorkappaB ligand/osteoclast differentiation factor in osteoclas-togenesis from synoviocytes in rheumatoid arthritis. ArthritisRheum 43:259–269

38. Emery JG, McDonnell P, Burke MB, Deen KC, Lyn S,Silverman C, Dul E, Appelbaum ER, Eichman C, DiprinzioR, Dodds RA, James IE, Rosenberg M, Lee JC, Young PR

Breast Cancer Res Treat (2007) 105:7–16 15

123

(1998) Osteoprotegerin is a receptor for the cytotoxic ligandTRAIL. J Biol Chem 273:14363–14367

39. Van Poznak C, Cross SS, Saggese M, Hudis C, Panageas KS,Norton L, Coleman RE, Holen I (2006) Espression of os-teoprotegerin (OPG), TNF related apoptosis inducing ligand(TRAIL), and receptor activator of nuclear factor kappaBligand (RANKL) in human breast tumours. J Clin Pathol59:56–63

40. Pederson L, Winding B, Foged NT (1999) Identification ofbreast cancer cell line-derived paracrine factors that stimu-late osteoclast activity. Cancer Res 59:5849–5855

41. Lau YS, Sabokbar A, Giele H, Cerundolo V, Hofstetter W,Athanasou NA (2006) Malignant melanoma and boneresorption. Br J Cancer 94:1496–503

16 Breast Cancer Res Treat (2007) 105:7–16

123