Loading

Annals of Breast Cancer Research

RANK or RANKL or OPG: The Axis of Breast Cancer Bone Metastasis Evil?

Review Article | Open Access

  • 1. Departmentof Biological Sciences, Texas Tech University, USA
  • 2. Department of Nutrition, Georgia State University, USA
  • 3. Department of Pathology, Texas Tech University Health Sciences Center, USA
+ Show More - Show Less
Corresponding Authors
Lauren S. Gollahon, Department of Biological Sciences, Texas Tech University, Lubbock, Texas 79409, USA, Tel: (806) 834-3287; Fax: (806) 742-2963
Abstract

Breast cancer bone metastasis remains the leading cause of death in individuals with advanced breast cancer of patients presenting with metastasis, ~ 70% is osseous. Generally, breast cancer bone metastasis is osteolytic, resulting in breakdown of the bone matrix. Treatment options are limited, and once breast cancer has metastasized to bone, it is incurable. The RANK/RANKL/OPG triad plays a critical role in breast cancer bone metastasis. Not only do the interactions of this triad facilitate and regulate normal bone turnover, it is also central to many signaling pathways in the metastatic bone microenvironment. Radiating from the RANK/RANKL/OPG axis, like spokes in a wheel, are factors such as TGF-beta, PTHrP, M-CSF, CCN2 and other cellular signaling pathways involved in the progression of breast cancer bone metastasis. Crosstalk between breast cancer cells and the bone microenvironment results in a vicious cycle of metastatic tumor promotion, progression and osteolysis. Furthermore, mounting evidence suggests that the RANK/RANKL/OPG axis also plays an important role in the development of breast carcinogenesis. However, the relationship between their differential expression in normal mammary epithelium and breast tumors is understudied and conflicting data from the literature muddies the role of each player in this triad in progression towards breast cancer bone metastasis. Thus, this review discusses the current status of RANK/RANKL/OPG expression and their involvement in breast cancer progression and metastasis and highlights the need to further understand the role of the triad.

Keywords

•    Breast cancer
•    Bone metastasis
•    RANKL
•    RANK
•    OPG

Citation

Schuster C, Mo H, Shen CL, Gollahon L (2017) RANK/RANKL/OPG: The Axis of Breast Cancer Bone Metastasis Evil? Ann Breast Cancer Res 2(1): 1008.

ABBREVIATIONS

BCBM: Breast Cancer Bone Metastasis; Casr: Calcium-Sensing Receptor; CCN2: CCN Family Protein 2; CREB: cAMP Response Element-Binding Protein; EMT: Epithelial To Mesenchymal Transition; ER: Estrogen Receptor; ERK: Extracellular SignalRegulated Kinase; IL-6: Interleukin-6; IL-8: Interleukin 8; JNK: C-Jun N-Terminal Kinase; M-CSF: Macrophage Colony-Stimulating Factor; MMP: Matrix Metalloproteinase; Nfatc1: Nuclear Factor of Activated T-Cells; OPG: Osteoprotegerin; PKA: Protein Kinase A; PKC: Protein Kinase C; PR: Progesterone Receptor; PTHrP: Parathyroid Hormone-Related Protein; RANK: Receptor Activator of Nuclear Factor Kappa-β; RANKL: Receptor Activator of Nuclear Factor Kappa-β Ligand; RRL O: RANK/RANKL/OPG; STAT5: Signal Transducer And Activator Of Transcription 5; TGF-β: Transforming Growth Factor-β; TNF: Tumor Necrosis Factor; TRAF6: TNF Receptor Associated Factor 6; TRAIL: TNFRelated Apoptosis-Inducing Ligand; VEGF: Vascular Endothelial Growth Factor

INTRODUCTION

Breast cancer is the most commonly diagnosed cancer in American women [1] and the second leading cause of cancerrelated deaths [2]. There are multiple risk factors associated with breast cancer, but the main two are gender and age [3]. Other risk factors include genetic predisposition, breast density, endogenous hormone levels, number of pregnancies, and use of oral contraceptives, obesity, and tobacco use and alcohol consumption [4]. Each year, over 200,000 new cases of invasive breast cancer and 60,000 new cases of in situ breast cancer are diagnosed in the United States [1]. Approximately 1 in 8 women in the United States will develop invasive breast cancer at least once during their life and 1 in 36 women will still die as a result of the disease [1], causing over 40,000 deaths.

Metastasis is the main cause of breast cancer-associated death [5] as localized disease is not considered fatal. Bone is the most common site to which breast cancer metastasizes. About 70% of breast cancers that metastasize spread to the bone [6]. 

Other sites for breast cancer metastases include lungs, liver and brain [5,7]. Five-year life expectancy decreases from about 95% for individuals with non-metastatic breast cancer to less than 25% for patients with metastatic breast cancer [8]. Once breast cancer has spread to the bone, it is deemed incurable [6]. Some of the current treatment options for bone metastases include radiotherapy and radiopharmaceuticals, orthopedic surgery, bisphosphonates, endocrine and cytotoxic treatments [9], most of which have drawbacks due to unfavorable side effects. Thus, understanding the underlying mechanisms in the pathogenesis and particularly the protein interactions governing the etiology of breast cancer-derived bone metastasis is critical for effectively preventing and treating the disease through the development of more targeted treatments, resulting in increased survivorship and quality of life. One such network may involve the Receptor Activator of Nuclear Factor Kappa-B (RANK)/ Receptor Activator of Nuclear Factor Kappa-B Ligand (RANKL)/Osteoprotegerin (OPG) (RRL O) axis, yet relatively little is known about expression, interactions and mechanisms of action of this signaling pathway in breast cancer metastasizing to the bone. Here, we review the current understanding of the RRL O triad in development of breast cancer and bone metastases and discuss the need for further examination of this understudied area that may yield a critical clue as to why bone is preferentially targeted by breast cancer metastasis.

The Chronology of Breast Cancer Bone Metastasis (BCBM)

Breast cancer cells have an arsenal of tools to facilitate escape from the confines of the primary tumor microenvironment. These include restructuring the extracellular matrix [10,11] and secreting proteases [11] and angiogenic factors [12], allowing the cancer cells to invade into circulation and become metastatic. Metastatic breast cells then release cytokines and growth factors [13-16] that promote adhesion in osseous tissue and subsequent proliferation [17]. While osseous breast cancer metastasis can occur in any bone, it most often occurs in bones proximal to the body midline, especially the spine [18]. Initially, circulating breast cancer cells preferentially adhere to the bone marrow endothelium [19].

Under normal circumstances, bone matrix is constantly fluctuating to maintain homeostasis through osteoblast - osteoclast activity. Osteoblasts are derived from mesenchymal cells that synthesize collagen fibers [20]. Once surrounded by newly deposited bone matrix, the osteoblasts differentiate into osteocytes. Osteoclasts are derived from a hematopoietic cell lineage [21]. Through release of proteolytic enzymes and lowering the extracellular pH, osteoclasts are able to resorb the bone matrix by dissolving hydroxyapatite crystals, subsequently releasing calcium and phosphate [22].

Once breast cancer cells have invaded the bone marrow, the balance between osteoblast and osteoclast activity is disrupted and the interaction between the dynamic bone environment and breast cancer cells facilitates the development of bone metastasis. Bone metastasis has been shown to induce higher rates of osteoclast activity [23,24]. Moreover, osteoblast differentiation is inhibited and apoptosis is induced [24]. This altered osteoclast

- osteoblast dynamism is facilitated by the secretion of growth factors and cytokines produced by breast cancer cells such as parathyroid hormone-related protein (PTHrP) [25], interleukin-6 (IL-6) [26], interleukin-8 (IL-8) [27], macrophage colonystimulating factor (M-CSF) [28], matrix metalloproteinase’s (MMPs) [29-32], and tumor necrosis factor (TNF) [33]. Secretion of these proteins plays an important role in facilitating metastatic behavior by directly or indirectly influencing osteoblasts and osteoclasts as well as their precursors. This in turn leads to higher osteoclast differentiation which leads to an increase in the secretion of growth factors and cytokines such as transforming growth factor-β (TGF-β) by osteoclasts which stimulates further breast cancer growth [34]. This results in a recursive loop that facilitates further metastasis and the pathogenic deterioration of bone integrity.

Molecular Mechanisms of BCBM: The Role of RRLO

The molecular mechanisms surrounding BCBM are very complex. It remains unclear how the RRL O triad, a signaling pathway most commonly known for its role in normal bone remodeling, regulates BCBM. RANK is a type I transmembrane protein and a member of the TNF receptor family [35,36]. RANKL is a type II transmembrane protein that belongs to the TNF super family [36]. OPG is a glycoprotein and decoy receptor to RANKL. It is also a member of the TNF receptor super family [37,38]. OPG, RANK and RANKL are widely expressed in many tissues including skin, smooth muscles, respiratory, gastrointestinal, and lymphoid tissues [38-41]. Interestingly, RRL O also has an observed role in mammary gland development [42], which may help elucidate its possible importance in BCBM and as potential biomarkers and prognostic factors should be changed to as a potential biomarker and a prognostic factor.

The Role of RRLO Interactions in Mammary Gland Development

Since the role of RRL O interactions is well known in bone development (See [43] for review), it will only be discussed in this review in context of possible links to BCBM. The importance of the relationship of RRL O in mammary gland development is much less understood and will be discussed below.

RANKL-RANK signaling has been shown to affect the expansion of mammary stem cells during normal mammary gland development [44,45]. Additionally, RANK and RANKL play an important role in development of the lactating mammary gland during pregnancy. Using a RANK/RANKL mouse knockout model, Fata et al., showed that lobuloalveolar development was impaired during pregnancy. This impairment was linked to a deficiency in the differentiation, proliferation, and survival of mammary epithelial cells [46]. In addition, RANKL expression in mammary cells is mainly regulated by progesterone and prolactin expression [47-50]. RANKL appears to be the main paracrine effector of progesterone for inducing mammary cell proliferation and expansion, and the resulting mammary gland development [44, 45,47,49,51-54]. Progesterone induces the proliferation of mammary epithelial cells through the progesterone receptor (PR) via a paracrine signaling mechanism mediated from PR-positive to neighboring PR-negative cells [55]. RANKL was shown to bind to the PR in a hormone-dependent manner [55]. This occurs when progesterone recruits signal transducer and activator of transcription 5 (STAT5) to complex with RANKL upon PR binding [55] (Figure 1).

An overview of the role of the RANK/RANKL/OPG axis in mammary gland development, tumorigenesis, and metastasis.

Figure 1: An overview of the role of the RANK/RANKL/OPG axis in mammary gland development, tumorigenesis, and metastasis.

RANKL and RANK both appear to have roles in mammary gland development, mammary epithelial and stem cell proliferation, differentiation and survival. In contrast, the role and mechanism of action of OPG remains unclear. This is in part, due to the paucity of studies investigating its function in normal breast epithelium. Based on its function as a decoy receptor to RANKL and its regulation of RANKL and RANK signaling [56,57], the involvement of OPG in regulating mammary gland development is also highly probable. Furthermore, OPG was observed in mammary epithelial cells [42]. Similarly, another study found the presence of OPG in human milk samples with levels of OPG more than 1000-fold greater than in normal human serum [58]. Since OPG is significantly raised in human milk samples postpartum, it suggests that not only RANKL-RANK signaling but also OPG might play a role in breast development during lactation. In contrast, using OPG knock-out mice, Mizuno et al, did not observe any abnormalities in the mouse mammary gland development, suggesting that OPG does not play a direct role in mammary gland development [59]. It is also possible that OPG might be inversely correlated with mammary epithelial cell proliferation [60].

RRLO in Breast Cancer

Increasing evidence suggests a vital role of the RRL O axis in tumor formation, progression and metastasis. Many studies show RANKL contributes directly to breast cancer formation and metastasis [53,54,61,62]. A major cancer-associated pathway regulated by RANKL-RANK binding is the NF-κB pathway. This pathway is activated in breast cancer cells that form tumor spheres (spheres formed by NF-κB activation in epithelial to mesenchymal transition (EMT), up regulation of interleukin-1B and interleukin-6 as well as cancer stem cells) [63]. A study by Palafox et al., found that RANKL lead to cancer progression and metastasis in mammary epithelial cells by inducing EMT and stemness [64]. Furthermore, several studies have also shown that an inhibition of RANKL or PR-associated pathways suppresses mammary tumor formation in mice [45,53,54,65].

However, when evaluating RANK, RANKL, and OPG expression at a transcriptional level, inconsistencies between studies and cell lines become apparent. For example, many breast cancer cell lines analyzed have been found to lack RANKL on the mRNA level with the exception of HCC70, while OPG and RANK seem to be expressed by most breast cancer cell lines [42,66- 69]. Several studies have also shown a lack of RANKL expression in primary and metastatic breast tumors, suggesting a possible down regulation in RANKL expression during the transformation from normal mammary epithelium to breast cancer. However, studies by Nicolin et al., found that MCF-7 and MDA-MB-231 breast cancer cells express RANKL protein, despite other studies finding a lack of RANKL mRNA expression [69-71]. Another study analyzed breast tumor tissues of patients and found that RANKL was expressed in 78.4% of patients and was associated with lower cell proliferation and improved skeletal disease-free survival [72].

While results from these studies do not paint a clear picture of the landscape for RANKL expression, even less is known about RANK expression. RANK is expressed in breast cancer cells lines such as MDA-MB-231, MCF-7, Hs578T, and ZR75-1 [73,74]. Santini et al., found that lower RANK mRNA expression in primary breast cancer specimens is associated with a longer overall survival [75].

Given the important and intimate interactions between the RRL O triad members, surprisingly little attention has been given to OPG in comparison to RANKL. One study reported that OPG was expressed in only 45.9% of breast tumor samples and its expression was associated with a smaller tumor size, node negativity, and lower cell proliferation [72]. Another study found 55% of primary breast tumors sampled expressed OPG, while it was highly expressed in mammary epithelium with columnar alteration. It was also expressed in endothelial cells but was absent in non-malignant lobules and ducts as well as myoepithelial cells [76]. This study further demonstrated that OPG expression is positively correlated with ER/PR status [76]. Another study found that increased OPG mRNA levels analyzed in primary breast cancer specimens correlate with longer overall survival [75]. A recent study investigated BRCA mutations and their correlation with OPG serum levels. Results showed that women post menopause with BRCA1/2 mutations have lower OPG serum levels than those without the mutation. Interestingly, OPG levels were inversely correlated with mammary epithelial proliferation [60]. However, since only serum levels were analyzed, it cannot be accurately predicted if we see the same correlation with OPG levels within the mammary tissue.

The RRLO Axis in Normal Bone Remodeling

RANK is expressed on precursors of osteoclasts. Binding of its ligand, RANKL, initiates the differentiation of osteoclast precursors into mature osteoclasts through the activation of the NF-κB pathways [36,39,77]. This results in the upregulation of Nuclear Factor of Activated T-cells (NFATc1), which is the master transcriptional regulator for osteoclast differentiation [78]. OPG is also expressed by osteoblasts. Once bound to RANKL, OPG inhibits RANK/RANKL interactions [36] and down regulates osteoclast differentiation [36,79] (Figure 2a).

The RANK/RANKL/OPG axis at the core of 2a) the normal bone remodeling vs 2b) the vicious cycle of breast cancer signaling leading to osteolysis.

Figure 2: The RANK/RANKL/OPG axis at the core of 2a) the normal bone remodeling vs 2b) the vicious cycle of breast cancer signaling leading to osteolysis.

Thus, a larger RANKL/OPG ratio expressed by osteoblasts is an indicator of an increased rate of osteoclastogenesis. A study by Nelson et al. showed that OPG binds to RANKL with an affinity that is approximately 500 fold higher than the binding affinity of RANK to RANKL [35].

A protein recently identified to interact with the RRL O triad, and possibly have a role in BCBM, is the CCN family protein 2 (CCN2). CCN2 is expressed in mesenchymally- derived cells including chondrocytes and osteoblasts. One study found that CCN2 bound to RANK in pre-osteoclastic RAW264.7 cells, inducing signaling pathways known to be RANK-induced such as NF-κB, p38 and Jun amino-terminal kinases (JNK) pathways. However, CCN2 did not impact RANKL-RANK binding and interactions [80]. Furthermore, CCN2 was also shown to bind to OPG, with a binding affinity close to that of OPG and RANKL. More intriguing was the finding that OPG inhibited the binding of CCN2 to RANK, suggesting that OPG might inhibit osteoclastogenesis not only by inhibiting RANKL to RANK binding, but also by suppressing CCN2 to RANK binding. Furthermore, the authors reported that CCN2 is an inhibitor of OPG’s negative regulation of osteoclastogenesis, although CCN2 does not directly inhibit RANKL to OPG binding. Therefore the authors suggest that the binding site of OPG and CCN2 is distinct from that for OPG and RANKL [80] (Figure 2a).

RRLO in Breast Cancer Bone Metastasis

Currently, the precise mechanism of action of RRL O in promoting metastasis remains unclear. MDA-MB-231 cells that expressed RANK have shown to have a higher metastatic growth rate than MDA-MB-231 cells that were RANK-negative [81]. A study by Santini et al., showed that RANK was positively correlated with the development of bone metastases and significantly higher expressed in estrogen receptor (ER) negative tumors [82]. Trinkaus et al., found that RANK was not expressed in normal breast tissue or primary breast cancer tissue of breast cancer patients, whereas 75% of patient samples expressed RANK in tumor cells at lymph nodes. However, the sample size was small (4 patients). They further showed that 50% of breast tumor cells at the metastatic bone site expressed RANK (sample size = 20) [83]. Another study found RANK to be expressed in 74.1% of patient breast tumor samples and was associated with a poor disease-free survival rate [72]. In contrast, Bhatia et al. showed no change in RANK expression among non-neoplastic breast tissue, non-metastatic infiltrating ductal carcinoma, metastatic infiltrating ductal carcinoma, and breast cancer bony metastases tissue [84]. To date, it remains unclear from in vivo studies [66,72,74,85] whether RANK should be considered a prognostic biomarker for BCBM.

RANKL was shown to induce markers for metastasis such as matrix metalloproteinase-1(MMP1) and vascular endothelial growth factor (VEGF) in MDA-MB-231 breast [73,85]. Several studies suggest that RANKL promotes breast cancer metastasis by directly affecting cancer stem cells or tumor cells that express RANK [62,86,87]. Protein expression from patient samples of normal breast tissue, primary and secondary (bone) metastasized breast tumors presents inconsistencies in expression levels. RANKL expression was shown to be down regulated from normal breast tissue samples to primary breast cancer tissue to breast cancer at the site of bone metastasis in two studies [76,84]. Bhatia et al., showed that RANKL expression was found in 90% of nonneoplastic breast tissue, 62% of non-metastatic infiltrating ductal carcinoma, 31% of metastatic infiltrating ductal carcinoma, and 2% of breast cancer bone metastases [84]. In contrast, another study found that all patient samples (n=4) expressed RANKL in the secondary breast tumor at the site of osseous metastasis [88]. Yet another in vitro study demonstrated that when the RANKLexpressing HCC70 breast cancer cell line was co-cultured with human osteoblasts, it decreased their RANKL expression [68].

The functional advantage of a possible decreased expression of RANKL in the primary and secondary tumor remains to be elucidated. One possibility for RANKL down regulation may be associated with the binding affinity of OPG to another ligand, TNFrelated apoptosis-inducing ligand (TRAIL), a ligand that binds to receptors on cells to facilitate apoptosis [89,90]. Interestingly, OPG has about equal binding affinities for both RANKL and TRAIL [91]. Some studies have proposed that the decreased RANKL expression in breast tumors and secondary tumors might be due to selective down regulation of RANKL by breast cancer cells, thus facilitating greater interactions between OPG and TRAIL, blocking TRAIL from binding to its receptor on breast cancer cells and consequently inhibiting apoptosis [76]. Holen et al., showed that OPG-expressing breast cancer cells efficiently inhibited TRAIL-induced apoptosis [92]. In contrast, Weichhaus et al., did not find an effect on TRAIL-mediated apoptosis upon OPG knockdown and suggested that there is another, as yet unknown mechanism involved in OPG’s role in metastasis and possibly downregulation of RANKL [37].

Several studies also found an association between an increase in OPG and a poorer prognosis in breast cancer through increased metastasis and invasion [37,69]. Knocking down OPG expression in MDA-MB-231 significantly reduced metastasis in the chick embryo metastasis model [37]. Moreover, OPG knockdown cells showed decreased invasion through collagen, an MMP-2 substrate [37], suggesting a role for OPG in breast cancer cell migration.

Taken together, OPG is upregulated and RANKL is down regulated during tumor progression and metastasis as an increase in OPG in breast cancer cells correlates with an increase in metastasis [37,69]. Further studies need to evaluate the roles of RANKL, RANK, and OPG as regulators of bone metastasis in human breast tissue and tumors as well as potential biomarkers and therapeutic targets.

The RRLO Triad at the Core of Many Signaling Pathways in Breast Cancer Bone Metastasis

As discussed above, OPG is significantly raised in human milk samples postpartum [58] and RANKL-RANK signaling plays a role in the mammary development during lactation. Calcium trafficking may be an underlying link between breast and bone in regards to the triad based on the nature of mammary gland production of milk and the function of osseous tissue in calcium regulation. In the bone environment, RANKL-RANK signaling is found to trigger NFATc1, leading to osteoclast differentiation and increased bone Reabsorption and remodeling [93]. RANKL is also responsible for inducing a sustained low level calcium oscillation that promotes the nuclear import of NFATc1 by activating calcineurin-NFATc1, which is an auto regulatory feedback mechanism that enhances its own expression and activates, with the aid of c-fos, a set of genes that are vital in osteoclastogenesis [78]. Activation of NFATc1 transcription in osteoclasts requires the co-factors TNF receptor associated factor 6 (TRAF6), NF-κB and c-fos. TRAF6 is also induced by RANKL to stimulate a NFκB pathway, which increases expression of NFATc1 by binding to NF-κB binding element in the promoter region. Thus, both the calcium-calcineurin and the TRAF6 - NF-κB pathways are utilized to activate NFATc1 activity [78].

Calcium signaling also plays an important role in the breast. Calcium-sensing receptor (CaSR), the master regulator of calcium metabolism, is expressed in mammary epithelial cells as well as breast cancer cells [94]. CaSR becomes activated during lactation and decreases levels of PTHrP in the milk and circulation, which leads to an increase of calcium in the milk [94]. During lactation there is usually an increase in bone turnover and bone resorption which frees calcium from the bone needed for lactation. It is suggested that during that time osseous RANKL is upregulated and OPG is down regulated. One study treated lactating mice with OPG and found that bone loss was reduced during lactation which, however, did not lead to a change in milk production, milk calcium levels or calcium homeostasis, unless dietary calcium was restricted [95]. Interestingly, osteoclast number remained the same but osteoblast number decreased suggesting that during lactation osteoclast activity is required for increased osteoclast numbers [95].

One longstanding paradigm is that RANKL was primarily expressed by osteoblasts [96]. However, more recent research has shown that osteocytes are the major source of RANKL in cancellous bone remodeling [97]. However, since metastatic breast cancer cells adhere to sites of bone turnover, the following discussion involves osteoblast expression of RANKL specifically.

Breast cancer cells that have migrated to the bone environment have been shown to secrete factors that increase the expression of RANKL in osteoblasts. PTHrP is known to be activated in breast cancer cells [94] and bind to receptors on osteoblasts, inducing RANKL production. Currently the exact mechanism that regulates the increased expression of RANKL in osteoblasts is not known. However, one study using mouse osteoblastic cells demonstrated that PTHrP-induced RANKL expression is dependent upon the activation of cAMP response element-binding protein (CREB) and the protein kinase A (PKA) and NFAT pathways [67]. PTHrP not only indirectly increases osteoclastogenesis, but it has also been shown to inhibit osteoblastogenesis and facilitate tumor growth of MDA-MB-231 breast cancer cells [98] (Figure 2b).

M-CSF, much like PTHrP, is released by breast cancer cells in the bone microenvironment and also up regulates RANKL expression on bone stromal cells [28]. This subsequently leads to an increase of osteoclastogenesis, resulting in osteolysis. Unlike PTHrP, M-CSF is itself a regulator of osteoclastogenesis in the normal bone environment. M-CSF, expressed by osteoblasts, binds to the c-FMS receptor on osteoclast precursors and activates the c-FOS pathway within the osteoclast precursors, up regulating NFATc1 and enabling monocytes, the precursors of osteoclasts, to fuse and form multinucleated osteoclasts [99] (Figure 2b).

As mentioned above, CCN2 has been shown to interact with the RRL O triad. One study found that bone morphogentic protein 9 (BMP-9) inhibited the bone metastasis of breast cancer cells through the downregulation CCN2 [100]. Another study found that an anti-CCN2 antibody was able to inhibit the osteolytic bone metastasis that appears with CCN2 and PTHrP overproduction. They showed that PTHrP significantly upregulated CCN2 in MDA-MB-231 cells and that CCN2 expression was promoted by PTHrP via PKA-, protein kinase C (PKC)-, and extracellular signalregulated kinase (ERK)-mediated signaling [101]. Interestingly, CCN2 is also needed for TGF-β-related signaling pathways such as Erk1/2 and Smad1 [102] (Figure 2b).

TGF-β is another important factor in this destructive BCBM loop. TGF-β is expressed by osteoblasts and osteoclasts and is upregulated during osteoclastic resorption of osteolytic bone during metastases [34]. This subsequently leads to the secretion of factors such as PTHrP, MMPs, TNF, IL-6, and IL-8 by the tumor, further driving bone destruction at the site of tumor adherence to the bone [34]. TGF-β stimulates the production PTHrP through Smad signaling in breast cancer cells, which closes the loop of this vicious cycle [103]. Furthermore, TGF-β has been shown to be involved in the epithelial-mesenchymal transition, angiogenesis and immunosuppression. Thus, TGF-β is a possible weak link in the metastasis armor to target for cancer therapy since blocking TGF-β might break the chain [104,105] (Figure 2b).

DISCUSSION & CONCLUSION

Interaction between breast cancer cells and the bone microenvironment results in a vicious cycle of bone metastasis. This vicious cycle is promoted through many factors and signaling pathways. Factors such as RANK, RANKL, OPG, TGF-β, PTHrP, M-CSF, and possibly CCN2 activate signaling pathways in breast cancer cells as well as osteoblasts and osteoclasts that lead to the resorption of the bone matrix and further stimulation of a more aggressive tumor phenotype. The RRL O triad plays an important role in osteolytic bone metastasis progression and appears to be the center point from which many factors and signaling pathways radiate. While the function of the RRL O axis in osteoclastogenesis is better understood, its role and expression in breast cancer remains to be elucidated. Currently, it is still unclear whether or how altered expression of RANKL and OPG in breast cancer cells directly influences BCBM. Furthermore, signaling pathways leading up to differential expression of RANKL and OPG in osteoblasts, breast cancer and mammary tissue remains largely unknown. More research is needed to understand the nature of the crosstalk between breast cancer cells and the bone microenvironment with regards to the RRL O axis. Understanding these signaling mechanisms will ultimately lead to a more targeted therapy for breast cancer bone metastases.

PR+ mammary epithelial cells secrete RANKL. RANKL binding to RANK on mammary stem and epithelial cells leads to the expansion and proliferation of mammary epithelial cells promoting mammary gland development. The role of OPG has not yet been established. Cellular stress and mutation may cause mammary epithelial cells to undergo tumorigenesis as depicted above. When TRAIL binds to its receptor TR R1/R2, it causes the breast tumor cells to undergo apoptosis. However, TRAIL binding to OPG leads to the inhibition of apoptosis. RANKL binding to its receptor RANK on breast tumor cells causes an increase in proliferation, survival and EMT. However, RANKL expression was found to be decreased in primary breast tumor tissues. What happens when RANKL binds at higher rates to OPG needs clarification, as does its expression in primary breast tumor tissue. RANKL expression appears to further decrease as primary breast cancer cells migrate and metastasize to the bone. OPG expression of breast tumor cells at the secondary bone site needs further elucidation.

Breast cancer cells secrete certain cytokines and growth factors, such as PTHrP and M-CSF that drive osteolysis. PTHrP and M-CSF stimulate osteoblasts to increase the secretion of RANKL, which then binds at higher rates to its receptor RANK driving the differentiation of osteoclast precursors into mature osteoclasts. This leads to higher rates of bone resorption, which in turn leads to greater secretion of growth factors such as TGF-β. This stimulates breast cancer cells to further increase the production of PTHrP which continues the vicious cycle seen in breast cancer bone metastasis. PTHrP appears to influence pathways involving CREB, NFAT and PKA in osteoblasts, causing them to increase RANKL expression. CCN2, secreted by osteoblasts and chondrocytes shown in Figure (2a) might interact with the triad, as it has also been shown to bind to RANK as well as OPG. In the presence of breast cancer cells in the bone environment, osteoblasts also increasingly secrete M-CSF, which binds to the c-FMS receptor on osteoclast precursors, driving osteoclastogenesis.

REFERENCES

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016; 66: 7-30.

2. U.S. Cancer Statistics Working Group. United States Cancer Statistics: 1999-2013 Incidence and Mortality Web-based Report. Atlanta (GA); 2016.

3. Hulka BS, Moorman PG. Breast cancer: hormones and other risk factors. Maturitas. 2001; 38: 103-13; discussion. 113-116.

4. Hankinson S, Hunter D. Breast Cancer. In: Adami HH, D T, editors. Textbook of Cancer Epidemiology. New York: Oxford University Press. 2002; 301-339.

5. Weigelt B, Peterse JL, van ‘t Veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005; 5: 591-602.

6. Roodman GD. Mechanisms of bone metastasis. N Engl J Med. 2004; 350: 1655-1664.

7. Lin NU, Bellon JR, Winer EP. CNS metastases in breast cancer. J Clin Oncol. 2004; 22: 3608-3617.

8. Howlader N, Noone AM, Krapcho M, Neyman N, Aminou R, Waldron W, et al. SEER Cancer Statistics Review, 1975-2010, National Cancer Institute. Bethesda, MD. 2013.

9. Coleman RE. Management of bone metastases. Oncologist. 2000; 5: 463-470.

10. Cox TR, Erler JT. Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. Dis Model Mech. 2011; 4: 165-178.

11. DeClerck YA, Mercurio AM, Stack MS, Chapman HA, Zutter MM, Muschel RJ, et al. Proteases, extracellular matrix, and cancer: a workshop of the path B study section. Am J Pathol. 2004; 164: 1131-1139.

12. Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis in cancer. Vasc Health Risk Manag. 2006; 2: 213-219.

13. Kinder M, Chislock E, Bussard KM, Shuman L, Mastro AM. Metastatic breast cancer induces an osteoblast inflammatory response. Exp Cell Res. 2008; 314: 173-183.

14. Esquivel-Velázquez M, Ostoa-Saloma P, Palacios-Arreola MI, NavaCastro KE, Castro JI, Morales-Montor J, et al. The role of cytokines in breast cancer development and progression. J Interferon Cytokine Res. 2015; 35: 1-16.

15. Sosnoski DM, Krishnan V, Kraemer WJ, Dunn-Lewis C, Mastro AM. Changes in cytokines of the bone microenvironment during breast cancer metastasis. Int J Breast Cancer. 2012.

16. Chen YC, Sosnoski DM, Mastro AM. Breast cancer metastasis to the bone: mechanisms of bone loss. Breast Cancer Res. 2010; 12: 215.

17. David Roodman G. Role of stromal-derived cytokines and growth factors in bone metastasis. Cancer. 2003; 97: 733-738.

18. Abeloff MD, Armitage JO, Niederhuber JE, Kastan MB, McKenna WG. Abeloff’s clinical oncology. Churchill Livingstone Philadelphia. 2008.

19. Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: a fatal attraction. Nat Rev Cancer. 2011; 11: 411-425.

20. Neve A, Corrado A, Cantatore FP. Osteoblast physiology in normal and pathological conditions. Cell Tissue Res. 2011; 343: 289-302.

21. Ash P, Loutit JF, Townsend KM. Osteoclasts derived from haematopoietic stem cells. Nature. 1980; 283: 669-670.

22. Stenbeck G. Formation and function of the ruffled border in osteoclasts. Semin Cell Dev Biol. 2002; 13: 285-292.

23. Mundy GR. Bone remodeling and its disorders. Metab bone Dis. 1999; 263.

24. Mercer RR, Miyasaka C, Mastro AM. Metastatic breast cancer cells suppress osteoblast adhesion and differentiation. Clin Exp Metastasis. 2004; 21: 427-435.

25. Henderson MA, Danks JA, Moseley JM, Slavin JL, Harris TL, McKinlay MR, et al. Parathyroid hormone-related protein production by breast cancers, improved survival, and reduced bone metastases. J Natl Cancer Inst. 2001; 93: 234-237.

26. Ara T, Declerck YA. Interleukin-6 in bone metastasis and cancer progression. Eur J Cancer. 2010; 46: 1223-1231.

27. Bendre MS, Gaddy-Kurten D, Mon-Foote T, Akel NS, Skinner RA, Nicholas RW, et al. Expression of interleukin 8 and not parathyroid hormone-related protein by human breast cancer cells correlates with bone metastasis in vivo. Cancer Res. 2002; 62: 5571-5579.

28. Mancino AT, Klimberg VS, Yamamoto M, Manolagas SC, Abe E. Breast cancer increases osteoclastogenesis by secreting M-CSF and upregulating RANKL in stromal cells. J Surg Res. 2001; 100: 18-24.

29. Pivetta E, Scapolan M, Pecolo M, Wassermann B, Abu-Rumeileh I, Balestreri, et al. MMP-13 stimulates osteoclast differentiation and activation in tumour breast bone metastases. Breast Cancer Res. 2011; 13: 105.

30. Yoneda T, Sasaki A, Dunstan C, Williams PJ, Bauss F, De Clerck YA, et al. Inhibition of osteolytic bone metastasis of breast cancer by combined treatment with the bisphosphonate ibandronate and tissue inhibitor of the matrix metalloproteinase-2. J Clin Invest. 1997; 99: 2509-2517.

31. Mehner C, Hockla A, Miller E, Ran S, Radisky DC, Radisky ES. Tumor cell-produced matrix metalloproteinase 9 (MMP-9) drives malignant progression and metastasis of basal-like triple negative breast cancer. Oncotarget. 2014; 5: 2736-2749.

32. Davies KJ. The Complex Interaction of Matrix Metalloproteinases in the Migration of Cancer Cells through Breast Tissue Stroma. Int J Breast Cancer. 2014; 2014: 839094.

33. Hamaguchi T, Wakabayashi H, Matsumine A, Sudo A, Uchida A. TNF inhibitor suppresses bone metastasis in a breast cancer cell line. Biochem Biophys Res Commun. 2011; 407: 525-530.

34. Juárez P, Guise TA. Tgf-Beta pathway as a therapeutic target in bone metastases. Curr Pharm Des. 2010; 16: 1301-1312.

35. Nelson CA, Warren JT, Wang MWH, Teitelbaum SL, Fremont DH. RANKL employs distinct binding modes to engage RANK and the osteoprotegerin decoy receptor. Structure. 2012; 20:1971-1982.

36. Boyce BF, Xing L. Biology of RANK, RANKL, and osteoprotegerin. Arthritis Res Ther. 2007; 9: S1.

37. Weichhaus M, Segaran P, Renaud A, Geerts D, Connelly L. Osteoprotegerin expression in triple-negative breast cancer cells promotes metastasis. Cancer Med. 2014; 3:1112-1125.

38. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Lüthy R , et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997; 89: 309-319.

39. Wada T, Nakashima T, Hiroshi N, Penninger JM. RANKL-RANK signaling in osteoclastogenesis and bone disease. Trends Mol Med. 2006; 12: 17-25.

40. Yun TJ, Chaudhary PM, Shu GL, Frazer JK, Ewings MK, Schwartz SM , et al. OPG/FDCR-1, a TNF receptor family member, is expressed in lymphoid cells and is up-regulated by ligating CD40. J Immunol. 1998; 161: 6113-6121.

41. Tan KB, Harrop J, Reddy M, Young P, Terrett J, Emery J, et al. Characterization of a novel TNF-like ligand and recently described TNF ligand and TNF receptor super family genes and their constitutive and inducible expression in hematopoietic and non-hematopoietic cells. Gene. 1997; 204: 35-46.

42. Thomas RJ, Guise TA, Yin JJ, Elliott J, Horwood NJ, Martin TJ, et al. Breast cancer cells interact with osteoblasts to support osteoclast formation. Endocrinology. 1999; 140: 4451-4458.

43. Boyce BF, Xing L. Functions of RANKL/RANK/OPG in bone modeling and remodeling. Arch Biochem Biophys. 2008; 473: 139-146.

44. Asselin-Labat ML, Vaillant F, Sheridan JM, Pal B, Wu D, Simpson ER , et al. Control of mammary stem cell function by steroid hormone signalling. Nature. 2010; 465: 798-802.

45. Joshi PA, Jackson HW, Beristain AG, Di Grappa MA, Mote PA, Clarke CL , et al. Progesterone induces adult mammary stem cell expansion. Nature. 2010; 465: 803-807.

46. Fata JE, Kong YY, Li J, Sasaki T, Irie-Sasaki J, Moorehead RA, et al. The osteoclast differentiation factor osteoprotegerin-ligand is essential for mammary gland development. Cell. 2000; 103: 41-50.

47. Brisken C, Ayyannan A, Nguyen C, Heineman A, Reinhardt F, Tan J, et al. IGF-2 is a mediator of prolactin-induced morphogenesis in the breast. Dev Cell. 2002; 3: 877-887.

48. Mulac-Jericevic B, Lydon JP, DeMayo FJ, Conneely OM. Defective mammary gland morphogenesis in mice lacking the progesterone receptor B isoform. Proc Natl Acad Sci USA. 2003; 100: 9744-9749.

49. Tanos T, Sflomos G, Echeverria PC, Ayyanan A, Gutierrez M, Delaloye JF, et al. Progesterone/RANKL is a major regulatory axis in the human breast. Sci Transl Med. 2013; 5:182.

50. Srivastava S, Matsuda M, Hou Z, Bailey JP, Kitazawa R, Herbst MP, et al. Receptor activator of NF-kappaB ligand induction via Jak2 and Stat5a in mammary epithelial cells. J Biol Chem. 2003; 278: 46171-46178.

51. Beleut M, Rajaram RD, Caikovski M, Ayyanan A, Germano D, Choi Y, et al. Two distinct mechanisms underlie progesterone-induced proliferation in the mammary gland. Proc Natl Acad Sci USA. 2010; 107: 2989-2994.

52. Fernandez-Valdivia R, Mukherjee A, Creighton CJ, Buser AC, DeMayo FJ, Edwards DP, et al. Transcriptional response of the murine mammary gland to acute progesterone exposure. Endocrinology. 2008; 149: 6236-6250.

53. Gonzalez-Suarez E, Jacob AP, Jones J, Miller R, Roudier-Meyer MP, Erwert R, et al. RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis. Nature. 2010; 468: 103- 107.

54. Schramek D, Leibbrandt A, Sigl V, Kenner L, Pospisilik JA, Lee HJ, et al. Osteoclast differentiation factor RANKL controls development of progestin-driven mammary cancer. Nature. 2010; 468: 98-102.

55. Obr AE, Grimm SL, Bishop KA, Pike JW, Lydon JP, Edwards DP. Progesterone receptor and Stat5 signaling cross talk through RANKL in mammary epithelial cells. Mol Endocrinol. 2013; 27: 1808-1824.

56. Ostrowski MC. A new role for OPG: putting RANKL in its place. J Bone Miner Res. 2010; 25: 1905-1906.

57. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, Martin TJ. Modulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand families. Endocr Rev. 1999; 20: 345-357.

58. Vidal K, van den Broek P, Lorget F, Donnet-Hughes A. Osteoprotegerin in human milk: a potential role in the regulation of bone metabolism and immune development. Pediatr Res. 2004; 55: 1001-1008.

59. Mizuno A, Amizuka N, Irie K, Murakami A, Fujise N, Kanno T, et al. Severe osteoporosis in mice lacking osteoclastogenesis inhibitory factor/osteoprotegerin. Biochem Biophys Res Commun. 1998; 247: 610-615.

60. Widschwendter M, Burnell M, Fraser L, Rosenthal AN, Philpott S, Reisel D, et al. Osteoprotegerin (OPG), The Endogenous Inhibitor of Receptor Activator of NF-?B Ligand (RANKL), is Dysregulated in BRCA Mutation Carriers. E BioMedicine. 2015; 2: 1331-1339.

61. Pellegrini P, Cordero A, Gallego MI, Dougall WC, Muñoz P, Pujana MA, et al. Constitutive activation of RANK disrupts mammary cell fate leading to tumorigenesis. Stem Cells. 2013; 31: 1954-1965.

62. Dougall WC, Holen I, González Suárez E. Targeting RANKL in metastasis. Bonekey Rep. 2014; 3: 519.

63. Kendellen MF, Bradford JW, Lawrence CL, Clark KS, Baldwin AS. Canonical and non-canonical NF-κB signaling promotes breast cancer tumor-initiating cells. Oncogene. 2014; 33: 1297-1305.

64. Palafox M, Ferrer I, Pellegrini P, Vila S, Hernandez-Ortega S, Urruticoechea A, et al. RANK induces epithelial-mesenchymal transition and stemness in human mammary epithelial cells and promotes tumorigenesis and metastasis. Cancer Res. 2012; 72: 2879- 2888.

65. Poole AJ, Li Y, Kim Y, Lin SC, Lee WH, Lee EY. Prevention of Brca1- mediated mammary tumorigenesis in mice by a progesterone antagonist. Science. 2006; 314: 1467-1470.

66. Owen S, Ye L, Sanders AJ, Mason MD, Jiang WG. Expression profile of receptor activator of nuclear-κB (RANK), RANK ligand (RANKL) and osteoprotegerin (OPG) in breast cancer. Anticancer Res. 2013; 33: 199-206.

67. Park HJ, Baek K, Baek JH, Kim HR. The cooperation of CREB and NFAT is required for PTHrP-induced RANKL expression in mouse osteoblastic cells. J Cell Physiol. 2015; 230: 667-679.

68. Schubert A, Schulz H, Emons G, Grundker C. Expression of osteoprotegerin and receptor activator of nuclear factor-kappaB ligand (RANKL) in HCC70 breast cancer cells and effects of treatment with gonadotropin-releasing hormone on RANKL expression. Gynecol Endocrinol. 2008; 24: 331-338.

69. Ney J, Fehm T, Juhasz-Boess I, Solomayer E. RANK, RANKL and OPG Expression in Breast Cancer - Influence on Osseous Metastasis. Geburtshilfe Frauenheilkd. 2012; 72: 385-391.

70. Nicolin V, Bortul R, Bareggi R, Baldini G, Martinelli B, Narducci P. Breast adenocarcinoma MCF-7 cell line induces spontaneous osteoclastogenesis via a RANK-ligand-dependent pathway. Acta Histochem. 2008; 110: 388-396.

71. Nicolin V, Narducci P. Soluble TRAIL could enhance bone destruction acting on Rank-ligand in estrogen-independent human breast cancer cell line MDA-MB-231. Acta Histochem. 2010; 112:189-192.

72. Park HS, Lee A, Chae BJ, Bae JS, Song BJ, Jung SS. Expression of receptor activator of nuclear factor kappa-B as a poor prognostic marker in breast cancer. J Surg Oncol. 2014; 110: 807-812.

73. Casimiro S, Mohammad KS, Pires R, Tato-Costa J, Alho I, Teixeira R, et al. RANKL/RANK/MMP-1 Molecular Triad Contributes to the Metastatic Phenotype of Breast and Prostate Cancer Cells In Vitro. PLoS One. 2013; 8: 63153.

74. Tometsko M, Armstrong A, Miller R, Jones J, Chaisson M, Branstetter D, et al. RANK ligand directly induces osteoclastogenic, angiogenic, chemoattractive and invasive factors on RANK-expressing human cancer cells MDA-MB-231 and PC3. J Bone Miner Res. 2004; 19: 25.

75. Santini D, Schiavon G, Vincenzi B, Gaeta L, Pantano F, Russo A, et al. Receptor activator of NF-kB (rank) expression in primary tumors associates with bone metastasis occurrence in breast cancer patients. PLoS One. 2011; 6: 19234.

76. Van Poznak C, Cross SS, Saggese M, Hudis C, Panageas KS, Norton L, et al. Expression of osteoprotegerin (OPG), TNF related apoptosis inducing ligand (TRAIL), and receptor activator of nuclear factor kappaB ligand (RANKL). J Clin Pathol. 2006; 59: 56-63.

77. Bai YD, Yang FS, Xuan K, Bai YX, Wu BL. Inhibition of RANK/RANKL signal transduction pathway: a promising approach for osteoporosis treatment. Med Hypotheses. 2008; 71: 256-258.

78. Takayanagi H. The role of NFAT in osteoclast formation. Ann N Y Acad Sci. 2007; 1116: 227-237.

79. Ryser MD, Qu Y, Komarova SV. Osteoprotegerin in bone metastases: mathematical solution to the puzzle. PLoS Comput Biol. 2012; 8: 1002703.

80. Aoyama E, Kubota S, Khattab HM, Nishida T, Takigawa M. CCN2 enhances RANKL-induced osteoclast differentiation via direct binding to RANK and OPG. Bone. 2015; 73: 242-248.

81. Blake ML, Tometsko M, Miller R, Jones JC, Dougall WC. RANK expression on breast cancer cells promotes skeletal metastasis. Clin Exp Metastasis. 2014; 31: 233-245.

82. Santini D, Perrone G, Roato I, Godio L, Pantano F, Grasso D, et al. Expression pattern of receptor activator of NFκB (RANK) in a series of primary solid tumors and related bone metastases. J Cell Physiol. 2011; 226: 780-784.

83. Trinkaus M, Ooi WS, Amir E, Popovic S, Kalina M, Kahn H, et al. Examination of the mechanisms of osteolysis in patients with metastatic breast cancer. Oncol Rep. 2009; 21: 1153-1159.

84. Bhatia P, Sanders MM, Hansen MF. Expression of receptor activator of nuclear factor-kappaB is inversely correlated with metastatic phenotype in breast carcinoma. Clin Cancer Res. 2005; 11: 162-165.

85. Rucci N, Millimaggi D, Mari M, Del Fattore A, Bologna M, Teti A, et al. Receptor activator of NF-?B ligand enhances breast cancer-induced osteolytic lesions through upregulation of extracellular matrix metalloproteinase inducer/CD147. Cancer Res. 2010; 70: 6150-6160.

86. Jones DH, Nakashima T, Sanchez OH, Kozieradzki I, Komarova SV, Sarosi I, et al. Regulation of cancer cell migration and bone metastasis by RANKL. Nature. 2006; 440: 692-696.

87. Armstrong AP, Miller RE, Jones JC, Zhang J, Keller ET, Dougall WC, et al. RANKL acts directly on RANK-expressing prostate tumor cells and mediates migration and expression of tumor metastasis genes. Prostate. 2008; 68: 92-104.

88. Huang L, Cheng YY, Chow LTC, Zheng MH, Kumta SM. Tumour cells produce receptor activator of NF-?B ligand (RANKL) in skeletal metastases. J Clin Pathol. 2002; 55: 877-878.

89. Pitti RM, Marsters SA, Ruppert S, Donahue CJ, Moore A, Ashkenazi A. Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J Biol Chem. 1996; 271: 12687-12690.

90. Wiley SR, Schooley K, Smolak PJ, Din WS, Huang CP, Nicholl JK, et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 1995; 3: 673-682.

91. Vitovski S, Phillips JS, Sayers J, Croucher PI. Investigating the interaction between osteoprotegerin and receptor activator of NFkappaB or tumor necrosis factor-related apoptosis-inducing ligan... J Biol Chem. 2007; 282: 31601-31609.

92. Holen I, Cross SS, Neville-Webbe HL, Cross NA, Balasubramanian SP, Croucher PI, et al. Osteoprotegerin (OPG) expression by breast cancer cells in vitro and breast tumors in vivo--a role in tumour cell survival? Breast Cancer Res Treat. 2005; 92: 207-215.

93. Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H, et al. Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell. 2002; 3: 889-901.

94. Vanhouten JN, Wysolmerski JJ. The calcium-sensing receptor in the breast. Best Pract Res Clin Endocrinol Metab. 2013; 27: 403-414.

95. Ardeshirpour L, Dumitru C, Dann P, Sterpka J, Van Houten J, Kim W, et al. OPG treatment prevents bone loss during lactation but does not affect milk production or maternal calcium metabolism. Endocrinology. 2015; 156: 2762-2773.

96. Xiong J, Piemontese M, Onal M, Campbell J, Goellner JJ, Dusevich V, et al. Osteocytes, not Osteoblasts or Lining Cells, are the Main Source of the RANKL Required for Osteoclast Formation in Remodeling Bone. PLoS One. 2015; 10: e0138189.

97. Xiong J, O’Brien CA. Osteocyte RANKL: new insights into the control of bone remodeling. J Bone Miner Res. 2012; 27: 499-505.

98. Zheng L, Zhu K, Jiao H, Zhao Z, Zhang L, Liu M, et al. Pthrp expression in human mda-mb-231 breast cancer cells is critical for tumor growth and survival and osteoblast inhibition. Int J Biol Sci. 2013; 9: 830-841.

99. Ross FP. M-CSF, c-Fms, and signaling in osteoclasts and their precursors. Ann N Y Acad Sci. 2006; 1068: 110-116.

100. Ren W, Sun X, Wang K, Feng H, Liu Y, Fei C, et al. BMP9 inhibits the bone metastasis of breast cancer cells by downregulating CCN2 (connective tissue growth factor, CTGF) expression. Mol Biol Rep. 2014; 41: 1373-1383.

101. Shimo T, Kubota S, Yoshioka N, Ibaragi S, Isowa S, Eguchi T, et al. Pathogenic role of connective tissue growth factor (CTGF/CCN2) in osteolytic metastasis of breast cancer. J Bone Miner Res. 2006; 21: 1045-1059.

102. Nakerakanti SS, Bujor AM, Trojanowska M. CCN2 is required for the TGF-β induced activation of Smad1-Erk1/2 signaling network. PLoS One. 2011; 6: e21911.

103. Guise T. Examining the metastatic niche: targeting the microenvironment. Semin Oncol. 2010; 37: 2-14.

104. Chiechi A, Waning DL, Stayrook KR, Buijs JT, Guise T a, Mohammad KS. Role of TGF-β in breast cancer bone metastases. Adv Biosci Biotechnol. 2013; 4: 15-30.

105. Buijs JT, Stayrook KR, Guise TA. TGF-β in the Bone Microenvironment: Role in Breast Cancer Metastases. Cancer Microenviron. 2011; 4: 261-281.

Received : 21 Jan 2017
Accepted : 03 Feb 2017
Published : 04 Feb 2017
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X