Skip to main content
Erschienen in: Molecular Cancer 1/2014

Open Access 01.12.2014 | Short communication

Direct but not indirect co-culture with osteogenically differentiated human bone marrow stromal cells increases RANKL/OPG ratio in human breast cancer cells generating bone metastases

verfasst von: Chiara Arrigoni, Paola De Luca, Mara Gilardi, Sara Previdi, Massimo Broggini, Matteo Moretti

Erschienen in: Molecular Cancer | Ausgabe 1/2014

Abstract

Background

Bone metastases arise in nearly 70% of patients with advanced breast cancer, but the complex metastatic process has not been completely clarified yet. RANKL/RANK/OPG pathway modifications and the crosstalk between metastatic cells and bone have been indicated as potential drivers of the process. Interactions between tumor and bone cells have been studied in vivo and in vitro, but specific effects of the direct contact between human metastatic cells and human bone cells on RANKL/RANK/OPG pathway have not been investigated.

Findings

We directly co-cultured bone metastatic human breast cancer cells (BOKL) with osteo-differentiated human mesenchymal cells (BMSCs) from 3 different donors. BMSCs and BOKL were then enzymatically separated and FACS sorted. We found a significant increase in the RANKL/OPG ratio as compared to control, which was not observed in BOKL cultured in medium conditioned by BMSCs, neither in BOKL directly cultured with fibroblasts or medium conditioned by fibroblasts. Direct co-culture with osteo-differentiated BMSCs caused BOKL aggregation while proliferation was not affected by co-culture. To more specifically associate RANKL expression to osteogenic differentiation degree of BMSCs, we determined their osteogenic markers expression and matrix calcification relative to osteoblasts and fibroblasts.

Conclusions

In conclusion, our co-culture model allowed to demonstrate for the first time that direct contact but not paracrine interactions between human metastatic breast cancer cells and bone cells has a significant effect on RANKL/OPG expression in bone metastatic cells. Furthermore, only direct contact with the bone microenvironment induced BOKL clustering without however significantly influencing their proliferation and migration.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-13-238) contains supplementary material, which is available to authorized users.
Chiara Arrigoni, Paola De Luca contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

CA drafted the manuscript, supervised cell cultures and participated in the design of experiments, PDL designed and performed cell cultures, PCR and analysis of time lapse data, MG helped with molecular biology experiments, SP executed time lapse analysis, MB and MM designed the study and critically revised the manuscript. All authors read and approved the final manuscript.
Abkürzungen
BMSCs
Bone marrow stromal cells
BOKL
MDA-MB231-BO-KL
CM
Conditioned medium
GM
Growth medium.

Findings

Introduction

Bone metastases arise in nearly 70% of patients with advanced breast cancer, indicating bone as a favorable microenvironment for metastases, which onset dramatically alters physiological bone turnover, leading to bone lysis. It has been reported [1] that this can be caused by the up-regulation of RANKL to OPG ratio, causing an increased osteoclastogenesis [2], although the mechanisms inducing RANKL/OPG modification are not clear [1, 3].
Recently, interactions between metastatic cells and metastatic niche cells have been indicated as potential drivers of the metastatic process [4]. Thus, to understand the cross-talk between metastatic breast cancer cells and bone microenvironment, many in vitro co-culture models have been established [510]. The majority of these [5, 6] involve human tumor cells cultured with mouse osteoblasts, but osteoblasts from different species have different characteristics [11] and mechanisms underlying metastasis formation can be diverse between mouse and human [12]. Moreover, while some studies reported the culture of human tumor cells in conditioned medium from human bone cells, highlighting only soluble factors effects [7, 8], only a few works [9, 10] implemented direct contact between human tumor and bone cells. However, gene expression analyses on individual cell populations have not always been performed [9].
Our work was thus aimed at investigating, by means of direct co-culture, the interactions between a line of fluorescently tagged bone metastatic human breast cancer cells, MDA-MB231-BO-KL (BOKL) and a bone microenvironment represented by osteo-differentiated primary human bone marrow stromal cells (BMSCs), in terms of modifications on RANKL/RANK/OPG expression, cell proliferation and migration.

Direct contact with bone-like cells induced RANKL/OPG up-regulation in metastatic cells

To investigate whether specific direct contact between bone and tumor cells is involved in RANKL/OPG ratio modifications, we compared gene expression of BOKL after 3 days of direct co-culture with osteo-differentiated BMSCs (co-culture BMSCs) with that of BOKL cultured in conditioned medium from osteo-differentiated BMSCs (CM BMSCs) and that of BOKL in direct or indirect co-culture with fibroblasts, non osteogenic cells (co-culture MRC-5, CM MRC-5). Based on literature data showing that cell populations with equivalent capacity to form bone metastasis also share equal expression of genes related to bone invasion and metastasis [13], we chose to use a single bone seeking clone [14], with equivalent bone metastatic potential as compared to other bone seeking clones [13]. BMSCs were harvested from 3 patients undergoing hip surgery, after informed consent, and were differentiated in osteogenic medium (OM), as described [15]. BOKL and BMSCs after direct co-culture with BMSCs or MRC-5 were retrieved by enzymatic digestion and separated by FACS sorting. To exclude modifications in gene expression caused by the separation procedure, gene expression levels were normalized respective to those of BOKL grown in GM and then subjected to the same separation procedure (control 1). CM for indirect co-culture was harvested from the same BMSCs and MRC-5 used in direct co-cultures and gene expression of BOKL in CM was normalized respective to BOKL grown in GM (control 2). All materials and methods are described in Additional file 1: Materials and methods.
Results of PCR analyses showed a strong and statistically significant upregulation (13 fold, p < 0.05) of RANKL in BOKL co-culture BMSCs as compared to control 1 but not in BOKL CM BMSCs and BOKL CM MRC-5 (Figure 1A, p < 0.05 and p < 0.01, respectively). A small increase (not statistically significant) in RANKL was observed in BOKL co-culture MRC-5 where a significant upregulation of OPG was found (Figure 1A), differently from all other conditions. Altogether our results show that RANKL/OPG ratio (a more useful indicator of effectively available RANKL) in BOKL co-culture BMSCs is 7 fold increased (Figure 1B) relative to control, and significantly higher (p < 0.01) than all other conditions, where no increase was found, demonstrating that direct, specific contact between tumor and bone cells can modify RANKL/OPG ratio. This suggests that among other factors hypothesized in the literature [1], heterotypic interactions between metastatic and bone cells can be involved in RANKL/OPG imbalance.
We demonstrated a specific activation of RANKL/OPG and not a generalized change in gene expression, since CDH11, known to be highly expressed in bone-seeking clones [16], and RANKL receptor RANK were not significantly modified in BOKL after direct or indirect co-cultures (Figure 1A). We also analyzed gene expression of BMSCs after direct co-culture and we found that average value of RANKL and osteogenic differentiation markers expression did not significantly differ between osteo-differentiated BMSCs before and after direct co-culture. However, 1 out of 3 patients derived osteo-differentiated BMSCs (patient 2) showed a 4-fold RANKL up regulation after direct co-culture, due to the high biological variability of primary human cells (Figure 1C, D). RANK expression was instead significantly upregulated in BMSCs after co-culture with tumor cells (Figure 1C, p < 0.01 as compared to BMSCs before co-culture), differently from MRC-5, which showed no expression of RANK before or after co-culture (data not shown), confirming a specific activation of the RANKL/OPG/RANK pathway caused by the crosstalk between tumor cells and bone microenvironment.
The separation of metastatic cells from bone-like cells was based on fluorescent labeling of cells before co-culture, giving a particular reliability to our results. Another study analyzed gene expression of tumor cells in direct co-culture with BMSCs [10], separating the two cell populations on the basis of epithelial markers expression. It is however known that highly aggressive breast cancer cells in co-culture with BMSCs can undergo epithelial-to-mesenchymal transition [17], causing a loss of epithelial markers and consequently affecting cell separation reliability.

Direct co-culture effects on BOKL aggregation, migration and proliferation

With the aim to analyze effects of direct contact between bone-like cells and BOKL on aggregation, migration and proliferation, time lapse analyses were performed, revealing an evident BOKL aggregation in direct co-culture with osteo-differentiated BMSCs, not noticeable in the other conditions, as shown in fluorescence images (Figure 2A, B, C, D). This phenomenon resembled cell filing, already described in pathological tissues bearing metastases [5]. Interestingly, BOKL clustering did not occur in co-culture with MRC-5, suggesting that this can represent a specific effect of the direct contact with bone cells. Cluster number normalized to total cell number was indeed significantly higher in BOKL co-culture BMSCs, as compared to other conditions (p < 0.001, Figure 2E). We observed a preferential direction of cell migration in direct co-culture as compared to culture with CM or GM (p < 0.01, Figure 2F), probably due to cell contact guidance [18] and not to specific characteristics of cells that composed the monolayer, being this effect evident also in co-culture with MRC-5. Figure 2G reports the quantification of BOKL proliferation in the different conditions, showing no significant differences between BOKL co-culture BMSCs and BOKL CM BMSCs. BOKL grown in 100% CM did not show a lower degree of proliferation as compared to control 2, indicating that CM can support BOKL growth. On the other hand, BOKL in contact with fibroblasts showed a significantly higher (p < 0.001) proliferation, accordingly to literature results showing how the release of specific factors (such as interleukin-6) by fibroblasts can increase the growth of breast cancer cells [19].

Characterization of bone-like microenvironment

To establish a bone-like environment we differentiated BMSCs for 14 days in osteogenic medium, having previous surveys and literature data [22] established that it allowed to achieve adequate osteo-differentiation. However, considering the time-dependence of differentiation markers expression and the different effects reported on tumor cells co-cultured with variously differentiated bone cells [6], we determined the osteodifferentiation degree of BMSCs used in our experiments with reference to both terminally differentiated osteoblasts and MRC-5. We characterized BMSCs osteodifferentiation through Alkaline Phosphatase (ALP) assay, Alizarin red staining and calcium quantification. Moreover, expression of both early (RUNX-2) and late osteogenic markers (osteopontin and osteocalcin) [23], was determined. After 14 days in osteogenic medium, BMSCs from all the 3 different donors showed positive Alizarin red staining (Figure 3A), demonstrating the presence of calcium deposits. Their quantification demonstrated significantly higher calcium levels in BMSCs as compared to MRC-5 (p < 0.001, Additional file 2: Figure S1A). ALP activity, index of initial osteogenic commitment of cells, normalized to total protein content, was detectable in all the 3 patients at comparable levels as shown in Additional file 2: Figure S1B. Moreover, the expression of osteogenic markers osteopontin, osteocalcin and RUNX-2 was significantly up-regulated in osteo-differentiated BMSCs as compared to MRC-5. Osteopontin and osteocalcin expression was significantly lower than in primary osteoblasts (p < 0.05) whereas expression of RUNX-2 was comparable (Figure 3B).
In conclusion, our simple co-culture model allowed to obtain results otherwise not possible to achieve in in vivo experiments, whereby it would be impossible to sort, separate and analyze the cell populations involved in the metastatic process. In particular, we demonstrated for the first time that direct but not indirect co-culture between human tumor cells and primary bone-like cells induced a significant up-regulation of RANKL/OPG expression in bone metastatic cells, suggesting an important role of direct contact in the bone metastatic process. This is in line with the reported clinical expression, determined by immunohistochemistry, of RANKL in human bone metastases [24, 25]. Furthermore, although we do not yet have a mechanistic explanation, upregulation of RANKL/OPG ratio and BOKL clustering observed only in direct co-culture with osteo-differentiated BMSCs and not in culture with conditioned medium or in direct co-culture with fibroblasts, further support the hypothesis that heterotypic cell interactions between tumor cells and bone cells can have a key role in the establishment of bone metastases.

Acknowledgements

This work was funded by the Italian Ministry of Health.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

CA drafted the manuscript, supervised cell cultures and participated in the design of experiments, PDL designed and performed cell cultures, PCR and analysis of time lapse data, MG helped with molecular biology experiments, SP executed time lapse analysis, MB and MM designed the study and critically revised the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Dougall WC: Molecular pathways: osteoclast-dependent and osteoclast-independent roles of the RANKL/RANK/OPG pathway in tumorigenesis and metastasis. Clin Cancer Res. 2012, 18: 326-335. 10.1158/1078-0432.CCR-10-2507CrossRefPubMed Dougall WC: Molecular pathways: osteoclast-dependent and osteoclast-independent roles of the RANKL/RANK/OPG pathway in tumorigenesis and metastasis. Clin Cancer Res. 2012, 18: 326-335. 10.1158/1078-0432.CCR-10-2507CrossRefPubMed
2.
Zurück zum Zitat Hofbauer LC, Schoppet M: Clinical implications of the osteoprotegerin/RANKL/RANK system for bone and vascular diseases. JAMA. 2004, 292: 490-495. 10.1001/jama.292.4.490CrossRefPubMed Hofbauer LC, Schoppet M: Clinical implications of the osteoprotegerin/RANKL/RANK system for bone and vascular diseases. JAMA. 2004, 292: 490-495. 10.1001/jama.292.4.490CrossRefPubMed
3.
Zurück zum Zitat Canon JR, Roudier M, Bryant R, Morony S, Stolina M, Kostenuik PJ, Dougall WC: Inhibition of RANKL blocks skeletal tumor progression and improves survival in a mouse model of breast cancer bone metastasis. Clin Exp Metastasis. 2008, 25: 119-129. 10.1007/s10585-007-9127-1CrossRefPubMed Canon JR, Roudier M, Bryant R, Morony S, Stolina M, Kostenuik PJ, Dougall WC: Inhibition of RANKL blocks skeletal tumor progression and improves survival in a mouse model of breast cancer bone metastasis. Clin Exp Metastasis. 2008, 25: 119-129. 10.1007/s10585-007-9127-1CrossRefPubMed
4.
Zurück zum Zitat Chaffer CL, Weinberg RA: A perspective on cancer cell metastasis. Science. 2011, 331: 1559-1564. 10.1126/science.1203543CrossRefPubMed Chaffer CL, Weinberg RA: A perspective on cancer cell metastasis. Science. 2011, 331: 1559-1564. 10.1126/science.1203543CrossRefPubMed
5.
Zurück zum Zitat Krishnan V, Shuman LA, Sosnoski DM, Dhurjati R, Vogler EA, Mastro AM: Dynamic interaction between breast cancer cells and osteoblastic tissue: comparison of two- and three-dimensional cultures. J Cell Physiol. 2011, 226: 2150-2158. 10.1002/jcp.22550CrossRefPubMed Krishnan V, Shuman LA, Sosnoski DM, Dhurjati R, Vogler EA, Mastro AM: Dynamic interaction between breast cancer cells and osteoblastic tissue: comparison of two- and three-dimensional cultures. J Cell Physiol. 2011, 226: 2150-2158. 10.1002/jcp.22550CrossRefPubMed
6.
Zurück zum Zitat Giunciuglio D, Cai T, Filanti C, Manduca P, Albini A: Effect of osteoblast supernatants on cancer cell migration and invasion. Cancer Lett. 1995, 97: 69-74. 10.1016/0304-3835(95)03955-VCrossRefPubMed Giunciuglio D, Cai T, Filanti C, Manduca P, Albini A: Effect of osteoblast supernatants on cancer cell migration and invasion. Cancer Lett. 1995, 97: 69-74. 10.1016/0304-3835(95)03955-VCrossRefPubMed
7.
Zurück zum Zitat Pohorelic B, Singh R, Parkin S, Koro K, Yang AD, Egan C, Magliocco A: Role of Src in breast cancer cell migration and invasion in a breast cell/bone-derived cell microenvironment. Breast Cancer Res Treat. 2012, 133: 201-214. 10.1007/s10549-011-1753-2CrossRefPubMed Pohorelic B, Singh R, Parkin S, Koro K, Yang AD, Egan C, Magliocco A: Role of Src in breast cancer cell migration and invasion in a breast cell/bone-derived cell microenvironment. Breast Cancer Res Treat. 2012, 133: 201-214. 10.1007/s10549-011-1753-2CrossRefPubMed
8.
Zurück zum Zitat Hsu YL, Hou MF, Kuo PL, Huang YF, Tsai EM: Breast tumor-associated osteoblast-derived CXCL5 increases cancer progression by ERK/MSK1/Elk-1/snail signaling pathway. Oncogene. 2013, 32: 4436-4447. 10.1038/onc.2012.444CrossRefPubMed Hsu YL, Hou MF, Kuo PL, Huang YF, Tsai EM: Breast tumor-associated osteoblast-derived CXCL5 increases cancer progression by ERK/MSK1/Elk-1/snail signaling pathway. Oncogene. 2013, 32: 4436-4447. 10.1038/onc.2012.444CrossRefPubMed
9.
Zurück zum Zitat Rajski M, Vogel B, Baty F, Rochlitz C, Buess M: Global gene expression analysis of the interaction between cancer cells and osteoblasts to predict bone metastasis in breast cancer. PLoS One. 2012, 7: e29743- 10.1371/journal.pone.0029743PubMedCentralCrossRefPubMed Rajski M, Vogel B, Baty F, Rochlitz C, Buess M: Global gene expression analysis of the interaction between cancer cells and osteoblasts to predict bone metastasis in breast cancer. PLoS One. 2012, 7: e29743- 10.1371/journal.pone.0029743PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Molloy AP, Martin FT, Dwyer RM, Griffin TP, Murphy M, Barry FP, O’Brien T, Kerin MJ: Mesenchymal stem cell secretion of chemokines during differentiation into osteoblasts, and their potential role in mediating interactions with breast cancer cells. Int J Cancer. 2009, 124: 326-332. 10.1002/ijc.23939CrossRefPubMed Molloy AP, Martin FT, Dwyer RM, Griffin TP, Murphy M, Barry FP, O’Brien T, Kerin MJ: Mesenchymal stem cell secretion of chemokines during differentiation into osteoblasts, and their potential role in mediating interactions with breast cancer cells. Int J Cancer. 2009, 124: 326-332. 10.1002/ijc.23939CrossRefPubMed
11.
Zurück zum Zitat Czekanska EM, Stoddart MJ, Richards RG, Hayes JS: In search of an osteoblast cell model for in vitro research. Eur Cell Mater. 2012, 24: 1-17.PubMed Czekanska EM, Stoddart MJ, Richards RG, Hayes JS: In search of an osteoblast cell model for in vitro research. Eur Cell Mater. 2012, 24: 1-17.PubMed
12.
Zurück zum Zitat Borowsky AD: Choosing a mouse model: experimental biology in context–the utility and limitations of mouse models of breast cancer. Cold Spring Harb Perspect Biol. 2011, 3: a009670-PubMedCentralCrossRefPubMed Borowsky AD: Choosing a mouse model: experimental biology in context–the utility and limitations of mouse models of breast cancer. Cold Spring Harb Perspect Biol. 2011, 3: a009670-PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, Guise TA, Massagué J: A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003, 3: 537-549. 10.1016/S1535-6108(03)00132-6CrossRefPubMed Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, Guise TA, Massagué J: A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003, 3: 537-549. 10.1016/S1535-6108(03)00132-6CrossRefPubMed
14.
Zurück zum Zitat Yoneda T, Williams PJ, Hiraga T, Niewolna M, Nishimura R: A bone-seeking clone exhibits different biological properties from the MDA-MB-231 parental human breast cancer cells and a brain-seeking clone in vivo and in vitro. J Bone Miner Res. 2001, 16: 1486-1495. 10.1359/jbmr.2001.16.8.1486CrossRefPubMed Yoneda T, Williams PJ, Hiraga T, Niewolna M, Nishimura R: A bone-seeking clone exhibits different biological properties from the MDA-MB-231 parental human breast cancer cells and a brain-seeking clone in vivo and in vitro. J Bone Miner Res. 2001, 16: 1486-1495. 10.1359/jbmr.2001.16.8.1486CrossRefPubMed
15.
Zurück zum Zitat Lopa S, Mercuri D, Colombini A, De Conti G, Segatti F, Zagra L, Moretti M: Orthopedic bioactive implants: hydrogel enrichment of macroporous titanium for the delivery of mesenchymal stem cells and strontium. J Biomed Mater Res A. 2013, 12: 3396-3403.CrossRef Lopa S, Mercuri D, Colombini A, De Conti G, Segatti F, Zagra L, Moretti M: Orthopedic bioactive implants: hydrogel enrichment of macroporous titanium for the delivery of mesenchymal stem cells and strontium. J Biomed Mater Res A. 2013, 12: 3396-3403.CrossRef
16.
Zurück zum Zitat Tamura D, Hiraga T, Myoui A, Yoshikawa H, Yoneda T: Cadherin-11-mediated interactions with bone marrow stromal/osteoblastic cells support selective colonization of breast cancer cells in bone. Int J Oncol. 2008, 33: 17-24.PubMed Tamura D, Hiraga T, Myoui A, Yoshikawa H, Yoneda T: Cadherin-11-mediated interactions with bone marrow stromal/osteoblastic cells support selective colonization of breast cancer cells in bone. Int J Oncol. 2008, 33: 17-24.PubMed
17.
Zurück zum Zitat Foroni C, Broggini M, Generali D, Damia G: Epithelial-mesenchymal transition and breast cancer: role, molecular mechanisms and clinical impact. Cancer Treat Rev. 2012, 38: 689-697. 10.1016/j.ctrv.2011.11.001CrossRefPubMed Foroni C, Broggini M, Generali D, Damia G: Epithelial-mesenchymal transition and breast cancer: role, molecular mechanisms and clinical impact. Cancer Treat Rev. 2012, 38: 689-697. 10.1016/j.ctrv.2011.11.001CrossRefPubMed
18.
Zurück zum Zitat Leong MC, Vedula SR, Lim CT, Ladoux B: Geometrical constraints and physical crowding direct collective migration of fibroblasts. Commun Integr Biol. 2013, 6: e23197- 10.4161/cib.23197PubMedCentralCrossRefPubMed Leong MC, Vedula SR, Lim CT, Ladoux B: Geometrical constraints and physical crowding direct collective migration of fibroblasts. Commun Integr Biol. 2013, 6: e23197- 10.4161/cib.23197PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Studebaker AW, Storci G, Werbeck JL, Sansone P, Sasser AK, Tavolari S, Huang T, Chan MW, Marini FC, Rosol TJ, Bonafè M, Hall BM: Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6-dependent manner. Cancer Res. 2008, 68: 9087-9095. 10.1158/0008-5472.CAN-08-0400CrossRefPubMed Studebaker AW, Storci G, Werbeck JL, Sansone P, Sasser AK, Tavolari S, Huang T, Chan MW, Marini FC, Rosol TJ, Bonafè M, Hall BM: Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6-dependent manner. Cancer Res. 2008, 68: 9087-9095. 10.1158/0008-5472.CAN-08-0400CrossRefPubMed
20.
Zurück zum Zitat Cserni G, Bianchi S, Vezzosi V, van Diest P, van Deurzen C, Sejben I, Regitnig P, Asslaber M, Foschini MP, Sapino A, Castellano I, Callagy G, Arkoumani E, Kulka J, Wells CA: Variations in sentinel node isolated tumour cells/micrometastasis and non-sentinel node involvement rates according to different interpretations of the TNM definitions. Eur J Cancer. 2008, 44: 2185-2191. 10.1016/j.ejca.2008.06.033CrossRefPubMed Cserni G, Bianchi S, Vezzosi V, van Diest P, van Deurzen C, Sejben I, Regitnig P, Asslaber M, Foschini MP, Sapino A, Castellano I, Callagy G, Arkoumani E, Kulka J, Wells CA: Variations in sentinel node isolated tumour cells/micrometastasis and non-sentinel node involvement rates according to different interpretations of the TNM definitions. Eur J Cancer. 2008, 44: 2185-2191. 10.1016/j.ejca.2008.06.033CrossRefPubMed
21.
Zurück zum Zitat Maheshwari G, Lauffenburger DA: Deconstructing (and reconstructing) cell migration. Microsc Res Tech. 1998, 43: 358-368. 10.1002/(SICI)1097-0029(19981201)43:5<358::AID-JEMT2>3.0.CO;2-DCrossRefPubMed Maheshwari G, Lauffenburger DA: Deconstructing (and reconstructing) cell migration. Microsc Res Tech. 1998, 43: 358-368. 10.1002/(SICI)1097-0029(19981201)43:5<358::AID-JEMT2>3.0.CO;2-DCrossRefPubMed
22.
Zurück zum Zitat Birmingham E, Niebur GL, McHugh PE, Shaw G, Barry FP, McNamara LM: Osteogenic differentiation of mesenchymal stem cells is regulated by osteocyte and osteoblast cells in a simplified bone niche. Eur Cell Mater. 2012, 23: 13-27.PubMed Birmingham E, Niebur GL, McHugh PE, Shaw G, Barry FP, McNamara LM: Osteogenic differentiation of mesenchymal stem cells is regulated by osteocyte and osteoblast cells in a simplified bone niche. Eur Cell Mater. 2012, 23: 13-27.PubMed
23.
Zurück zum Zitat Long MW: Osteogenesis and bone-marrow-derived cells. Blood Cells Mol Dis. 2001, 27: 677-690. 10.1006/bcmd.2001.0431CrossRefPubMed Long MW: Osteogenesis and bone-marrow-derived cells. Blood Cells Mol Dis. 2001, 27: 677-690. 10.1006/bcmd.2001.0431CrossRefPubMed
24.
Zurück zum Zitat Brown JM, Corey E, Lee ZD, True LD, Yun TJ, Tondravi M, Vessella RL: Osteoprotegerin and rank ligand expression in prostate cancer. Urology. 2001, 57: 611-616. 10.1016/S0090-4295(00)01122-5CrossRefPubMed Brown JM, Corey E, Lee ZD, True LD, Yun TJ, Tondravi M, Vessella RL: Osteoprotegerin and rank ligand expression in prostate cancer. Urology. 2001, 57: 611-616. 10.1016/S0090-4295(00)01122-5CrossRefPubMed
25.
Zurück zum Zitat Peng X, Guo W, Ren T, Lou Z, Lu X, Zhang S, Lu Q, Sun Y: Differential expression of the RANKL/RANK/OPG system is associated with bone metastasis in human non-small cell lung cancer. PLoS One. 2013, 8: e58361- 10.1371/journal.pone.0058361PubMedCentralCrossRefPubMed Peng X, Guo W, Ren T, Lou Z, Lu X, Zhang S, Lu Q, Sun Y: Differential expression of the RANKL/RANK/OPG system is associated with bone metastasis in human non-small cell lung cancer. PLoS One. 2013, 8: e58361- 10.1371/journal.pone.0058361PubMedCentralCrossRefPubMed
Metadaten
Titel
Direct but not indirect co-culture with osteogenically differentiated human bone marrow stromal cells increases RANKL/OPG ratio in human breast cancer cells generating bone metastases
verfasst von
Chiara Arrigoni
Paola De Luca
Mara Gilardi
Sara Previdi
Massimo Broggini
Matteo Moretti
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2014
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-13-238

Weitere Artikel der Ausgabe 1/2014

Molecular Cancer 1/2014 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.