Skip to main content
Erschienen in: Molecular Cancer 1/2010

Open Access 01.12.2010 | Research

Runx2 transcriptome of prostate cancer cells: insights into invasiveness and bone metastasis

verfasst von: Sanjeev K Baniwal, Omar Khalid, Yankel Gabet, Ruchir R Shah, Daniel J Purcell, Deepak Mav, Alice E Kohn-Gabet, Yunfan Shi, Gerhard A Coetzee, Baruch Frenkel

Erschienen in: Molecular Cancer | Ausgabe 1/2010

Abstract

Background

Prostate cancer (PCa) cells preferentially metastasize to bone at least in part by acquiring osteomimetic properties. Runx2, an osteoblast master transcription factor, is aberrantly expressed in PCa cells, and promotes their metastatic phenotype. The transcriptional programs regulated by Runx2 have been extensively studied during osteoblastogenesis, where it activates or represses target genes in a context-dependent manner. However, little is known about the gene regulatory networks influenced by Runx2 in PCa cells. We therefore investigated genome wide mRNA expression changes in PCa cells in response to Runx2.

Results

We engineered a C4-2B PCa sub-line called C4-2B/Rx2dox, in which Doxycycline (Dox) treatment stimulates Runx2 expression from very low to levels observed in other PCa cells. Transcriptome profiling using whole genome expression array followed by in silico analysis indicated that Runx2 upregulated a multitude of genes with prominent cancer associated functions. They included secreted factors (CSF2, SDF-1), proteolytic enzymes (MMP9, CST7), cytoskeleton modulators (SDC2, Twinfilin, SH3PXD2A), intracellular signaling molecules (DUSP1, SPHK1, RASD1) and transcription factors (Sox9, SNAI2, SMAD3) functioning in epithelium to mesenchyme transition (EMT), tissue invasion, as well as homing and attachment to bone. Consistent with the gene expression data, induction of Runx2 in C4-2B cells enhanced their invasiveness. It also promoted cellular quiescence by blocking the G1/S phase transition during cell cycle progression. Furthermore, the cell cycle block was reversed as Runx2 levels declined after Dox withdrawal.

Conclusions

The effects of Runx2 in C4-2B/Rx2dox cells, as well as similar observations made by employing LNCaP, 22RV1 and PC3 cells, highlight multiple mechanisms by which Runx2 promotes the metastatic phenotype of PCa cells, including tissue invasion, homing to bone and induction of high bone turnover. Runx2 is therefore an attractive target for the development of novel diagnostic, prognostic and therapeutic approaches to PCa management. Targeting Runx2 may prove more effective than focusing on its individual downstream genes and pathways.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-9-258) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

Conceived and designed the experiments: SKB and BF. Performed the experiments: SKB, OK, YG, AEG, SS and DJP. Bioinformatics analysis: RRS and DM. Analyzed data and wrote the paper: SKB, GAC and BF. All authors have read and approved the final manuscript.

Introduction

Runx2 together with Runx1 and Runx3 comprise the Runx class of transcription factors, defined by their highly homologous Runt-related DNA-binding domain. As heterodimers with Cbfß, Runx proteins bind to cognate DNA elements with the consensus nucleotide sequence 5'-ACCACA in the promoters/enhancers of their target genes [1]. The three Runx proteins coordinate proliferation and differentiation of various cell types [2]. Runx1 is important for hematopoiesis [3, 4]; Runx2 is pivotal in osteogenesis [1, 5, 6]; and Runx3 is critical for neurogenesis [7], thymopoiesis [4], and maintenance of the gastric epithelium [4, 8]. While promoting specific cellular phenotypes, Runx proteins have evolved to inhibit cell proliferation. Runx3 is a bona fide tumor suppressor [9] as down-regulation of its promoter by hypermethylation contributes to the development of gastric cancer [10, 11]. Ablation of Runx1 activity leads to leukemia [12] and disruption of Runx2 results in deregulated cell proliferation and immortalization [1317]. Paradoxically, Runx2 is also implicated in carcinogenesis. In a mouse screen for c-Myc-collaborating oncogenes, MLV-induced leukemia occurred most frequently when the provirus integrated into the Runx2 locus resulting in its ectopic expression [18]. It was suggested that Runx2 initially provides the cells with a survival advantage, and its anti-mitogenic activity is counteracted by the CD2-Myc transgene present in the mouse model used for this screen [2, 19]. Therefore, Runx2-mediated tumorigenesis likely requires additional loss of check-point genes such as Trp53 or improper regulation of an oncogene such as c-Myc [19].
Runx2 has been extensively studied in the context of osteoblastogenesis from mesenchymal progenitors, where as a master regulator it stimulates the expression of various bone matrix components such as osteocalcin and bone sialoprotein (BSP) [20]. Runx2-/- mice die soon after birth due to the lack of differentiated osteoblasts and thus a mineralized skeleton [1, 5, 6]. Runx2 haploinsufficiency in humans causes the rare skeletal disorder Cleidocranial Dysplasia [21]. In search for hints to explain the high predilection of prostate and breast cancer to metastasize to bone, investigators have noticed ectopic expression of Runx2 and some of its target genes in biopsies from advanced tumors and their derivative cell lines [2226]. In a mouse model of PCa, conditional deletion of Pten in prostate epithelial cells resulted in the development of tumors with progressive increase in Runx2 expression [27]. Among the osteomimetic properties of prostate and breast cancer cells are expression of the Runx2 target genes MMP9 [28], BSP [29] and VEGFA [30], as well as induction of mineralization [25].
In addition to promoting osteoblast differentiation, Runx2 drives the expression of osteoclastogenic signals, both in osteoblasts [31, 32] and in the PC3 bone metastasis-derived PCa cell line [22]. PC3 cells robustly express Runx2 [33], and its silencing decreased their osteoclastogenic property in vitro and their growth within the bone microenvironment in vivo [22]. Runx2 also promotes metastatic aspects not necessarily related to bone. Invasion of PC3 cells through Matrigel™, a basement membrane-like preparation, decreased after Runx2 silencing [22], and its ectopic expression in mammary epithelial cells increased their proliferation and disrupted their normal acinar organization [34]. An oncogenic role for Runx2 has also been suggested in tumors that do not exhibit high predilection to bone, including pancreatic ductal adenocarcinoma [35] and thyroid papillary carcinoma [36].
Whereas Runx2 is being increasingly recognized as a pro-metastatic factor, little is known about the underlying transcriptional programs. To establish gene regulatory networks downstream of Runx2 in aggressive PCa, we analyzed gene expression in response to Runx2 in the C4-2B PCa cell line. These cells are castration-resistant derivatives of the androgen-dependent LNCaP cells, and serve as a model for the aggressive stage of bone metastatic PCa [37, 38]. Although C4-2B cells express Runx2 at levels higher than LNCaP cells [25], these levels are far lower than those observed in PC3 cells or osteoblasts [22]. We therefore engineered a C4-2B sub-line that allowed us to profile gene expression after induction of Runx2 with Doxycycline to levels seen in PC3 cells. Remarkably, the most significant changes were the up-regulation of genes implicated in cancer progression and cellular movement, and the down-regulation of genes involved in cell cycle progression. Consistent with these changes in gene expression, Runx2 enhanced PCa cell invasiveness and inhibited their proliferation.

Results and Discussion

Establishment of C4-2B PCa cells with conditional Runx2 expression

To establish a C4-2B cell line that conditionally expresses Runx2, we employed the recently described lentivirus-based pSLIK vector system, which allows tight Doxycycline (Dox)-inducible, RNA PolII-mediated transcription of a gene of interest [39]. C4-2B cells were transduced with Flag-tagged Runx2-encoding lentiviruses (Figure 1A), resulting in the C4-2B/Rx2dox subline. As control, we established the C4-2B/Rx2-Mdox subline, where Dox treatment induced expression of the transcriptionally inactive Flag-Runx2-M (Figure 1A) [40]. Western blot analysis with anti-Flag antibodies confirmed roughly equal expression levels of the wild type and mutant Runx2 proteins, which were strictly and dose-dependently regulated by Dox (Figure 1B). RT-qPCR analysis revealed that the Dox treatment increased Runx2 mRNA by ~20-fold compared to its endogenous levels, and that the induced level was comparable to that observed in the PC3high sub-line (Figure 1C). Western analysis using anti-Runx2 antibodies indicated that the level of endogenous Runx2 protein was negligible in untreated C4-2B cells, and that Dox induced expression of the exogenous Runx2 to the levels normally found in osteoblasts (Figure 1D) [41].
The transcriptional activity of Dox-induced Runx2 was initially assessed using luciferase reporter assay (Figure 1E). In the reporter plasmid 6XOSE2-Luc, luciferase expression was controlled by six copies of the osteoblast-specific element 2 (OSE2) from the Runx2-regulated osteocalcin (OC) gene promoter [42]. In the absence of Dox, 6XOSE2-luc activity was indistinguishable from the background luciferase activity observed without any cell extract, suggesting lack of endogenous Runx2 activity (Figure 1E). The luciferase reporter was strongly stimulated by WT but not by the mutant form of Runx2 (Figure 1E). As shown in Figure 1F, Runx2 also stimulated transcription of its endogenous target genes Bone Sialoprotein (BSP) [29] and Matrix Metalloprotease-9 (MMP9) [28]. These genes were not stimulated in the Dox-treated C4-2B/Rx2-Mdox cells (data not shown). Interestingly, Runx2 did not significantly enhance the expression of OC (Figure 1F) and Alkaline Phophatase (ALP; data not shown), although these genes are strongly stimulated by Runx2 in osteoblasts [1, 43]. This observation reflects cell type-dependent Runx2-mediated transcriptional control, and is consistent with the results of Yeung et al. [33], who demonstrated that in PC3 cells the OC promoter is responsive to the transcription factors AP-1 and SP1, but not Runx2. To identify Runx2-regulated genes and pathways in advanced PCa cells in an unbiased manner, we subjected C4-2B/Rx2dox cells to global gene expression profiling.

Runx2-regulated global gene expression and in silico assessment of associated pathways

C4-2B/Rx2dox cells were subjected to microarray gene expression analysis after one- and two- days of treatment with either Dox or vehicle in biological quadruplicates (a total of 16 samples). Of 24,526 probes represented in the microarray, 532 genes showed ≥2-fold increased expression and 378 genes showed ≥2-fold decreased expression with high statistical significance (p < 0.008) on either day of treatment (see additional file 1). RT-qPCR analysis of 50 representative genes conformed to the microarray data (Table 1 and additional file 2).
Table 1
Genes most responsive to Runx2 in C4-2B cells
Gene ID
Symbol
Gene Name
Anova
Fold change
Description of cancer related or other major function
Reference
   
p value
I
II
q
  
Transcription regulators
7069
THRSP
Thyroid hormone responsive
3.5E-06
7
5
 
Transcription of enzymes involved in tumor lipid metabolism
 
29947
DNMT3L
DNA (cytosine-5-)-methyltransferase 3-like
5.5E-08
5
6
> 50
DNA methylation, nucleic acid metabolic process
 
4091
SMAD6
SMAD family member 6
8.1E-10
5
4
 
Cell communication, TGF-beta receptor signaling pathway
 
6591
SNAI2
Snail homolog 2 (Drosophila)
8.8E-07
4
12
6
Promotes EMT by repressing E-cadherin transcription
[44]
6662
SOX9
SRY (sex determining region Y)-box 9
4.4E-07
4
6
17
Promotes chondrocyte differentiation, cell migration, and EMT
[110]
7025
NR2F1
Nuclear receptor subfamily 2,
3.5E-07
3
14
2
Ligand-dependent receptor, regulation of transcription termination
 
678
ZFP36L2
Zinc finger protein 36, C3H type-like 2
4.1E-09
3
10
 
Regulation of RNA metabolic process
 
4088
SMAD3
SMAD family member 3
6.3E-10
3
7
25
Signal transducer downstream of TGF-β receptors
 
283248
RCOR2
REST corepressor 2
2.9E-06
3
7
 
Negative regulation of transcription
 
10217
CTDSPL
Small CTD phosphatase 3
1.9E-09
-2
-3
 
Phosphatase, dephosphorylates Rb, suppresses cellular growth
 
1870
E2F2
E2F transcription factor 2
8.2E-10
-2
-5
-4
Transcription activator, regulation of cell cycle progression, proliferation
 
90390
MED30
Mediator complex subunit 30
< 1.0E-12
-2
-3
 
Transcription activator, Androgen receptor signaling pathway
 
4609
MYC
V-myc myelocytomatosis viral oncogene
2.9E-07
-2
-3
-2
Positive regulation of cell proliferation, apoptosis
[111, 112]
29893
PSMC3IP
PSMC3 interacting protein
3.5E-08
-2
-4
 
Gene expression from RNA PolII
 
4602
MYB
Viral oncogene homolog (avian)
1.6E-08
-3
-5
-3
G1/S transition of mitotic cell cycle
 
4824
NKX3-1
NK3 homeobox 1
3.7E-08
-3
-3
 
Transcription repressor, loss induces prostate tumorigenesis
 
Intracellular signaling, cell cycle and proliferation
51655
RASD1
RAS, dexamethasone-induced 1
6.8E-12
26
19
> 50
Ras related, suppresses cell proliferation, inhibits tumor growth
[92]
6446
SGK
Serum/glucocorticoid regulated kinase 1
< 1.0E-12
19
45
 
Cellular Na+ ion homeostasis, cell survival
 
57007
CXCR7
Chemokine receptor 7
9.2E-11
8
12
30
Receptor for SDF-1, GPCR, enhanced integrin activation and tumor growth
[113]
1843
DUSP1
Dual specificity phosphatase 1
9.1E-13
8
8
10
Regulator of cell cycle, response to stress
[114]
5580
PRKCD
Protein kinase C, delta
2.0E-11
6
5
12
Serine-Threonine Kinase, cell proliferation, cell contraction
 
55890
GPRC5C
G protein-coupled receptor, family C
1.3E-09
5
5
25
GPCR, cell communication
 
10461
MERTK
C-mer proto-oncogene tyrosine kinase
6.2E-10
4
6
39
Protein amino acid phosphorylation
 
5920
RARRES3
Retinoic acid receptor responder 3
4.3E-08
3
8
40
Negative regulation of cell proliferation
 
1903
EDG3
Sphingosine-1-phosphate receptor 3
4.8E-10
3
7
 
GPCR, enhances tumor growth, cell migration, and angiogenesis
[7779, 115, 116]
8877
SPHK1
Sphingosine kinase 1
7.5E-09
3
12
36
Kinase for the production of S1P, enhances tumor growth, cell migration, and angiogenesis, PCa cell survival against chemotherapy, osteoclast chemotaxis
[7779, 115, 116]
7049
TGFBR3
Transforming growth factor, β receptor III
7.1E-09
3
4
12
Negative regulation of TGFβ receptor signaling pathway
 
4921
DDR2
Discoidin domain receptor tyrosine kinase 2
2.9E-05
2
5
 
Collagen binding, cell proliferation
 
1017
CDK2
Cyclin-dependent kinase 2
1.3E-03
-2
-3
-4
Kinase, mitotic cell cycle, cell proliferation
 
55038
CDCA4
Cell division cycle associated 4
7.2E-08
-2
-4
-4
Repressor of E2F-dependent transcriptional activation and cell proliferation
 
5997
RGS2
Regulator of G-protein signaling 2, 24 kDa
9.0E-09
-3
-3
 
Cell cycle, GPCR signaling pathway, cell communication
 
25805
BAMBI
BMP and activin membrane-bound
inhibitor homolog (Xenopus laevis)
9.7E-10
-2
-3
 
TGF-β signaling pathway
 
Cellular movement, adhesion, invasion, cytoskeleton remodeling
89797
NAV2
Neuron navigator 2
6.7E-09
9
10
29
Scaffold protein, actin cytoskeleton remodeling, nervous system development
[53]
27111
SDCBP2
Syndecan binding protein 2
2.2E-03
6
15
 
Binds SDC2, regulates cell adhesion and communication with ECM molecules
[84, 85]
11344
PTK9L
Twinfilin
9.0E-10
6
5
11
Actin-binding protein, cytoskeleton organization
[57]
83715
ESPN
Espin
2.1E-08
5
8
> 50
Actin-binding-bundling protein, actin cytoskeleton dynamics, cell locomotion
[54, 117, 118]
51474
LIMA1
Eplin, LIM domain and actin binding 1
3.0E-10
4
5
15
Cytoskeleton-associated protein, regulation of actin filament depolymerization
[56]
8829
NRP1
Neuropilin 1
4.1E-08
4
11
40
TM, coreceptor for VEGF, collagen binding, cell adhesion, invasion
[119]
4582
MUC1
Mucin 1, cell surface associated
1.1E-09
4
4
 
TM, cell adhesion, invasion
[120]
4131
MAP1B
Microtubule-associated protein 1B
7.1E-09
3
9
18
Trans-membrane glycoprotein, cytoskeleton organization
[55, 121]
9644
SH3PXD2A
SH3 and PX domains 2A
1.4E-09
3
7
 
Adaptor protein, ECM degradation by actin based invadopedia formation
[122]
6275
S100A4
S100 calcium binding protein A4
2.6E-08
2
24
> 50
EF-hand calcium-binding protein, promotes cell motility, tumor metastasis
[49]
6383
SDC2
Syndecan 2
3.1E-06
2
5
32
TM, bind signaling factors, cell adhesion, angiogenesis, ECM degradation
[84, 85]
1013
CDH15
Cadherin 15, type 1, M-cadherin
6.9E-08
2
5
21
TM, Ca2+dependent intercellular adhesion, skeletal muscle development
[123]
Secreted factors
1437
CSF2
Colony stimulating factor 2/GM-CSF
2.3E-09
12
17
> 50
Cytokine, osteolytic bone metastasis
[72]
5225
PGC
Progastricsin (pepsinogen C)
3.9E-12
11
15
 
Aspartyl proteinase
[47]
5304
PIP
Prolactin-induced protein
< 1.0E-12
10
79
> 50
Aspartyl proteinase, ligand for CD4 receptor T cells
[46]
8857
FCGBP
Fc fragment of IgG binding protein
1.5E-09
5
9
 
Cell adhesion, cell-cell recognition
 
6387
CXCL12
Chemokine ligand 12 (SDF-1)
2.2E-06
5
7
23
Chemokine, stem cell trafficking to bone, migration, metastasis of tumor cells
[5865]
7422
VEGFA
Vascular endothelial growth factor A
4.5E-10
5
3
4
Growth factor, angiogenesis, cellular morphogenesis during differentiation
[8891]
1907
EDN2
Endothelin 2
4.3E-07
5
2
 
Growth factor, vasoactive, G-protein signaling, smooth muscle contraction
[87]
5654
HTRA1
HtrA serine peptidase 1
6.9E-10
3
18
 
Serine protease, differentially expressed in osteoarthritic cartillage
[124]
3817
KLK2
Kallikrein-related peptidase 2
1.5E-08
4
3
 
Serine protease, protein metabolic process
 
4880
NPPC
Natriuretic peptide precursor C
1.8E-10
4
17
> 50
Peptide, potent natriuretic, diuretic, and vasodilating
 
7057
THBS1
Thrombospondin 1
3.0E-10
4
14
> 50
Cell adhesion, heparin-binding, anti-angiogenic
 
3934
LCN2
Lipocalin 2
5.1E-08
4
10
5
ECM protein, present ligands to cell surface receptors, promotes EMT
[45]
6462
SHBG
Sex hormone-binding globulin
8.7E-10
4
10
 
Steroid-binding, signal
 
57642
COL20A1
Collagen, type XX, α 1
4.9E-07
4
10
> 50
Adhesion, establishment of localization, transport
 
4254
KITLG
KIT ligand
2.3E-08
4
7
41
Hematopoietic growth factor and ligand for the KIT tyrosine kinase receptor
 
4884
NPTX1
Neuronal pentraxin I
2.5E-07
3
20
 
Cell-cell signaling, transport, establishment of localization
 
3486
IGFBP3
IGF binding protein 3
2.2E-08
3
7
 
Regulation of cell growth, communication, apoptosis
 
9806
SPOCK2
Sparc/osteonectin, cwcv, kazal-like
domains proteoglycan (testican) 2
1.1E-06
3
4
 
Cell differentiation, ECM organization and biogenesis
 
8530
CST7
Cystatin F (leukocystatin)
3.0E-09
2
25
 
Competitive inhibitors of cysteine proteases, metastasis associated protein
[48]
4885
NPTX2
Neuronal pentraxin II
7.6E-09
2
6
 
Cell-cell signaling, synaptic transmission
 
147381
CBLN2
ADAM MMP with Thbs-1 motif
5.2E-12
-3
-3
 
Peptidase, cleavage of precerebellin in the nervous system
 
6781
STC1
Spondin 2
3.6E-08
-3
-3
 
ECM protein, integrin ligand, pattern recognition
 
7035
TFPI
Tissue factor pathway inhibitor
3.9E-05
-3
-4
 
Kunitz-type inhibitor of blood coagulation by blocking factor Xa
 
9518
GDF15
Growth differentiation factor 15
1.6E-09
-2
-8
-7
Growth factor, TGF-β signaling pathway, cell communication
 
Fold changes on day 1 and day 2 are listed in columns I and II respectively, and column q lists fold changes based on RT-qPCR analysis of the day-2 samples. Major gene functions were determined using knowledge databases from OMIM (Online Mendelian Inheritance of Man), IPA™ systems and the indicated referenced literature. Abbreviations: EMT, epithelial to mesenchymal transition; TM, transmembrane; GPCR, G-protein coupled receptor; ECM, extracellular matrix.
An unsupervised hierarchical analysis of these 910 up- and down-regulated genes resulted in a clear separation between the Dox-treated and control samples (Figure 2). The variation among the biological quadruplicates was small, indicating the overall robustness of the methodology utilized. Gene clusters showing changes in expression pattern with respect to time and Dox treatment were clearly discernable. In general, changes observed on day 1 of treatment were maintained or intensified by day 2.
We next employed the Ingenuity Pathway Analysis (IPA™) platform to indentify disease pathways, as well as molecular and cellular functions associated with Runx2-regulated genes. The analysis suggested 'cancer' as the disease most significantly associated with both the up- and the down-regulated gene groups (Table 2). A total of 248 genes, half from each group, had highly significant cancer related function (see additional file 3). Additionally, the up-regulated genes were strongly associated with genetic disorders, inflammatory responses, and gastrointestinal diseases (Table 2A). Among the most significant molecular and cellular functions, cellular movement, cell death, cellular growth, and proliferation were associated with the up-regulated genes, whereas cell cycle, cell death, cellular assembly and DNA replication functions were associated with the down-regulated genes (Table 2B).
Table 2
Diseases, Molecular and Cellular Functions associated with genes up-regulated (A) and down-regulated (B) by Runx2
A) ≥ 2 fold up-regulated, total 532 genes
p-value
genes
   Diseases and disorders
  
Cancer
2.5e-07 - 3.0e-03
124
Genetic disorder
5.4e-07 - 3.0e-03
229
Inflammatory response
3.3e-06 - 2.4e-03
65
   Molecular and cellular functions
 
Cellular movement
9.7e-12 - 2.5e-03
84
Cell death
2.6e-10 - 3.2e-03
129
Cellular growth, proliferation
9.3e-08 - 3.2e-03
108
B) ≥ 2 fold down-regulated, total 378 genes
 
   Diseases and disorders
 
Cancer
3.3e-29 - 4.1e-03
124
Gastrointestinal disease
3.3e-29 - 3.7e-03
72
Genetic disorder
3.3e-06 - 3.4e-03
174
   Molecular and cellular functions
 
Cell cycle
7.1e-35 - 4.1e-03
102
Cellular assembly, organization
1.7e-18 - 3.4e-03
63
DNA replication, recombination, repair
1.7e-18 - 4.1e-03
101
IPA™ analysis indicating the association of Runx2-regulated genes to a particular category identifying the indicated disease or function. The ranges of p values indicate the statistical significance with which Runx2-regulated genes associate with the each category.

Runx2-modulated genes are involved in tumor metastasis

Promotion of tissue invasion, metastasis and cytoskeleton dynamics

The major functions reported for the up-regulated genes belonged to cancer progression (Table 2). Importantly, these genes encode transcriptional regulators, cytoskeletal components, signaling molecules and peptidases, which have been implicated in tumor metastasis (Table 1). The transcription factors Sox9 and SNAI2, and the extracellular-matrix (ECM) protein LCN2, all major regulators of epithelial-to-mesenchymal transition (EMT) [44, 45], were up-regulated by ~4-fold after one day and by > 6-fold after two days of Runx2 induction, and their upregulation was confirmed by RT-qPCR (Table 1). However, the functional significance of these EMT markers requires further investigation in light of the unexpected increase in E-cadherin mRNA (see additional file 4). Runx2 also enhanced the expression levels of multiple transcripts encoding matrix modifying peptidases (Table 1). These included MMP9, a known Runx2 target in BCa cells [28] and aspartyl proteases with fibronectin degrading activities such as Prolactin Induced Protein (PIP) and Pepsinogen (PGC) [46, 47]. The latter two showed a rapid ~10-fold increase within 24 hours (Table 1) and PIP exhibited the highest change (79-fold) in response to Runx2 on day 2 (Table 1 and additional file 2). PIP protein in the C4-2B/Rx2dox culture supernatant was below detectable levels under control conditions, but was readily detected after induction of Runx2 (Figure 3). We also found increased transcript levels for Cystatin 7 (CST7), S100A4 and SMAD3, with a mild ~3-fold increase on day 1, but a robust > 20-fold increase on day 2 (Table 1). These genes function as metastasis promoters [48, 49]. Interestingly, S100A4 and SMAD3 physically interact to potentiate cancer cell invasiveness [50].
Runx2 also up-regulated genes involved in cellular movement and cytoskeleton remodeling (Table 1). SH3PXD2A, which was up-regulated by 7-fold on day 2, is a scaffold protein involved in the formation of invadopedia [51, 52], which are matrix digesting, actin rich, short lived protrusions observed in osteoclasts and cancer cells [53]. Runx2 up-regulated by > 9-fold the transcripts for Nav2, a scaffold protein crucial for actin cytoskeleton remodeling [53]. Other genes that were up-regulated by > 3-fold, with known roles in actin cytoskeleton dynamics included ESPN, which interacts with the Src-homology 3 (SH3) adaptor proteins to regulate cytoskeletal actin functions [54]; MAP1B, known to maintain cytoskeletal integrity [55]; LIMA1, which cross-links actin monomers [56]; and PTK9L (a.k.a. twinfiln), which sequesters ADP-actin monomers in the cytoplasm and delivers them to sites of rapid actin-filament assembly [57].

Metastasis to bone and modification of the bone microenvironment

The expression of SDF-1 and its receptor CXCR7 was enhanced by > 5-fold based on the microarray analysis and by > 20-fold based on the RT-qPCR results (Table 1). Runx2-induced SDF-1 protein was also detectable in the culture supernatant (Figure 3). SDF-1 signaling is critical for homing of hematopoietic cells to the bone marrow space and their survival in this environment [5865]. Within one day, Runx2 also increased by 10-fold the mRNA for BSP (Figure 1F), whose abundant expression by bone metastatic tumor cells facilitates their attachment to the bone matrix [6669]. Once settled in the bone microenvironment, the metastatic cells secrete regulatory molecules that stimulate bone turnover [70]. Remarkably, Runx2 enhanced the expression of the osteoclastogenic cytokine CSF2 by > 50-fold within 48 hours (Table 1). This presumably occurred by direct binding of Runx2 to the CSF2 promoter [71]. Runx2-mediated induction of CSF2 in PCa cells likely contributes to the increased bone turnover in bone metastatic sites, similar to the role of this cytokine in breast cancer bone metastasis [72]. CSF2 production by tumor cells may also contribute to accumulation of macrophages, inflammatory T cells, and cytokines [73, 74] that exacerbate morbidity and mortality [75]. Two additional Runx2-up-regulated genes associated with osteoclast function are SPHK1, a kinase responsible for the production of sphingosine 1 phosphate (S1P), and S1P receptor 3 (S1P3) a.k.a. EDG3 (Table 1). Production of S1P in the bone microenvironment promotes bone resorption by chemotactically attracting osteoclast precursors [76]. The SPHK1/S1P/S1P3 axis plays additional roles in cancer progression, including cell growth, migration, angiogenesis, and resistance to chemotherapy [7779]. Notably, Runx2 was the only gene differentially up-regulated in chemotherapy-resistant versus -sensitive osteosarcoma tumors [80]. Adding to this, Runx2 also repressed the expression of GDF-15, an osteoclastogenesis inhibitor [81] (Table 1). This repression was mild on day 1 (2-fold), but by day 2 GDF-15 was the most repressed gene in response to Runx2 (8-fold; Table 1 and additional file 2). Thus, Runx2-mediated alterations in gene expression may contribute to both the predilection of PCa to bone and the subsequent pathological increase in bone turnover, which further fuels growth of the metastatic tumors.

Angiogenesis

Runx2 has been implicated in promoting angiogenesis by stimulating VEGFA expression during bone development [30] as well as during tumorigenesis [82]. In C4-2B/Rx2dox cells, Runx2 increased VEGFA mRNA by 4-fold (Table 1) and the presence of VEGF in the cell culture supernatant was detectable only after Dox treatment (Figure 3). Additionally, our study revealed a 32-fold upregulation of the VEGFA co-receptor Syndecan-2 (SDC2; Table 1). SDC2, which is also a Runx2 target in osteoprogenitor cells [83], is a member of the heparan sulfate proteoglycans family, and is also implicated in cell adhesion and communication [8486]. Interestingly, VEGFA can functionally synergize with SDF-1 to promote neoangiogenesis in vivo [62]. Our microarray analysis also revealed Runx2-mediated induction of the angiogenic EDN-2 gene (Table 1). Endothelins and VEGFA are secreted by PCa cells to stimulate angiogenesis as well as differentiation of neighboring osteoblasts in the bone microenvironment [72, 8791].

Runx2 increases the invasion potential of C4-2B cells in vitro

Because Runx2 enhanced the expression of multiple extracellular enzymes involved in ECM degradation (Table 1), we initially tested by in-gel zymography the presence of proteases in the supernatant of Dox-treated C4-2B/Rx2dox cultures. The results demonstrated that Runx2 induced several gelatin-degrading proteins, in particular one with a molecular weight of ~140 kDa, the identity of which remains to be determined (Figure 4A). We further investigated whether Runx2 stimulates invasion of C4-2B/Rx2dox cells through Matrigel™, a tissue basement membrane-like preparation containing laminin, type IV collagen, heparan sulfate proteoglycans and entactin. For convenient and accurate assessment of cells that successfully invade through the Matrigel™ membrane, we transduced the C4-2B/Rx2dox cells with a lentivirus constitutively expressing luciferase. Parallel trans-wells that do not contain Matrigel™ were employed as migration controls. Cells were incubated in the respective chambers in the presence or absence of Dox, and the relative migration or invasion capacity was assessed. Runx2 expression led to a 2.3-fold decrease in cell migration, but a 4.3-fold increase in invasion through Matrigel™, i.e., a ~10-fold increase in invasiveness after adjustment for the reduced cell migration (Figure 4B). The increased invasiveness was further confirmed in an independent experiment by histological staining (Figure 4C). In parallel experiments, expression of Runx2-M in C4-2B cells showed no significant effects on either migration or invasion (data not shown). Thus, expression of transcriptionally active Runx2 is sufficient to enhance the tissue invasion potential of C4-2B cells.

Runx2 induces cellular quiescence by reversibly inhibiting G1/S cell cycle transition

The IPA™ analysis of Runx2 down-regulated genes indicated their strong association with cell cycle and proliferation-related functions (Table 2B). Many of these down-regulated genes, as well as several up-regulated genes, shed light on the well-established anti-proliferative activity of Runx2 [2, 1517, 19]. Most striking was the > 19- and > 8-fold up-regulation of RASD1 and DUSP1, respectively (Table 1). RASD1 belongs to the Ras superfamily of G-proteins, and its expression in breast cancer suppressed cell growth [92]. DUSP1, a.k.a. MAP kinase phosphatase 1 (MKP1), is a dual specificity (Thr/Tyr) protein phophatase with anti-proliferative properties [93]. Among the most important cell cycle regulatory genes inhibited by Runx2 was c-Myc, with a ~3-fold decrease at the mRNA level (Table 1) and a corresponding significant decrease at the protein level (Figure 3). In line with the down-regulation of c-Myc, the mRNA encoding its cell cycle promoting targets E2F2 and CDK2 were also down-regulated (Table 1). CDK2 protein was decreased below detectability (Figure 3). To further characterize effects of Runx2 on PCa cell proliferation, we first validated by RT-qPCR the changes in the transcript levels of RASD1, DUSP1, c-Myc and E2F2 in the day-2 samples (Figure 5A). Next, we tested the effect of Runx2 on C4-2B cell proliferation by performing MTT assays every 48 hours after Dox-mediated Runx2 induction. Runx2 significantly restrained cell proliferation (Figure 5B). By contrast, the transcriptionally-inactive Runx2-M did not affect proliferation (Figure 5C). Thus, Runx2 restrains PCa cell proliferation via its transcriptional activation property.
To delineate the anti-proliferative effect of Runx2, we tested its influence on apoptosis and cell cycle progression. Apoptosis was measured using succinyl-AMC, a fluorogenic Caspase-3 substrate. Expression of Runx2, but not Runx2-M, resulted in a transient 40% increase in apoptosis on day 4 (Figure 5D), but this could not account for the dramatic inhibition of cell proliferation (Figure 5B). Instead, fluorescence activated cell-sorting (FACS) analysis revealed a 2.2-fold decrease in the fraction of cells in the S/G2/M phases of the cell cycle within 24 hours of Dox treatment, and this effect persisted on day 2 (Figure 5E). Notably, the cell cycle inhibition preceded any change in apoptosis, indicating that Runx2 restrained C4-2B cell proliferation by inhibiting the G1/S phase transition of the cell cycle.
The prometastatic but anti-mitogenic properties of Runx2 in PCa cells suggest that it may initially facilitate metastasis, after which it must be degraded or antagonized (e.g. by co-repressors) to allow cell proliferation and tumor growth. Therefore, we examined if the anti-mitogenic effect of Runx2 was reversible by withdrawing Dox from cultures after 48 hours of treatment (Figure 5F). Dox withdrawal led to substantial clearance of Runx2 within two days, and undetectable Runx2 levels after four days (Figure 5G). This resulted in resumption of cell cycle progression (Figure 5H) and cell proliferation (Figure 5I). Thus, the Runx2-regulated gene networks induce reversible cellular quiescence by blocking the G1/S phase transition of the cell cycle.

Generality and Network modeling of Runx2-regulated genes with cancer-related functions

Our study is the first to provide genome wide analysis of Runx2-regulated genes in PCa cells. Although we focused on the bone metastasis-derived C4-2B cells, similar responses to Runx2 were observed in the parental lymph node-derived LNCaP cells [38] (see additional file 5) as well as in the unrelated bone metastatic 22RV1 PCa cells (see additional file 6) [94]. Furthermore, in PC3high and PC3low cells with high and low levels of Runx2, respectively, the expression of six randomly selected Runx2 up-regulated genes from the present study correlated with Runx2 expression (see additional file 7). Together, these results suggest that our observations are relevant to various stages of PCa progression. Among the 910 genes that Runx2 up- or down-regulated by ≥2 fold, IPA™ identified 248 genes related to cancer with high statistical significance (p < 0.003; Table 2 and additional file 3). The IPA™ analysis, as well as survey of literature on gene expression profiling in osteoblasts and fibroblasts, further suggested that the Runx2-regulated gene network in PCa (Figure 6) bears little resemblance to its targets in mesenchymal cells [43, 83, 9597]. In fact, only five of the cancer-related genes in our study have been previously reported as Runx2 targets (MMP13, CEBPA, VEGF, SMAD3, and SMAD6) and only six others are Runx1 and/or Runx3 targets (SKP2, CSF2, IL3, ACHE, IGFBP3, and HIPK2; Figure 6). The remaining 234 genes are therefore novel Runx2-regulated genes related to cancer in general and to metastasis in particular. The extensive Runx2-regulated cancer-related gene network highlights Runx2 as a viable target for the diagnosis, prognosis and treatment of PCa.

Conclusions

Runx2, traditionally known for its master regulatory roles in the chondro-osteoblast lineage, is emerging as a prometastatic transcription factor [22, 29, 34, 98101]. The Runx2 transcriptome in C4-2B cells documents gene networks that control multiple aspects of metastasis. Potentially contributing to local invasion and dissemination are the genes known to function in EMT, motility and ECM degradation. Additionally, the prometastatic function of Runx2 likely involves its target genes SDF-1, CXCR7 and BSP, which promote homing and attachment to bone. We also discovered Runx2 targets such as CSF2 and SPHK1, osteoclast activators that likely contribute to the most important alteration that occurs in the bone microenvironment in response to PCa metastasis, namely enhanced bone turnover. During this process, bone matrix components such as TGFß, BMPs and calcium ions are released and further fuel tumor growth and bone microenvironment modifications [102105]. The regulation of SPHK1 by Runx2 probably potentiates additional aspects of the cancer phenotype, including angiogenesis (a function assisted by the Runx2 targets VEGFA and EDN2) and drug resistance [77, 79].
The anti-mitogenic activity of Runx2 is consistent with the slow growth of PCa tumors, and may contribute to drug resistance. We imagine that future anti-Runx2 drugs will be administered along with traditional chemotherapy to eliminate cells that regain proliferative capacity. Interestingly, Runx2 physically and functionally interacts with the receptors for androgens and estrogens [99, 101]. Since these receptor proteins are targeted by many drugs for prostate and breast cancer, it is important to investigate their effects on Runx2-regulated transcription. Moreover, development of selective estrogen and androgen receptor modulators may benefit from consideration of their effects on Runx2 and expression of its target genes reported in the present study.

Methods

Cell culture reagents and antibodies

C4-2B cells were obtained from ViroMed Laboratories (Minneapolis, MN). LNCaP and 22RV1 cells were from ATCC (Rockville, MD, USA). PC3 cells were also obtained from ATCC, but propagated for several years in either our laboratory (PC3low) or that of USC's Dr. Pradip Roy-Burman (PC3high). The cells were maintained in RPMI-1640 medium supplemented with 10% Tet System Approved FBS from Clontech, CA, USA. Hygromycin B was purchased from Invitrogen, Carlsbad, CA, USA and added to the growth medium at 50 μg/ml. Doxycycline from Calibiochem, La Jolla, CA, USA was used at 0.25 ug/ml unless otherwise stated, and an equal volume of distilled water was used as vehicle control. Mouse ANTI-FLAG® M2 monoclonal antibody for was purchased from Sigma, St Louis, MO, USA. Mouse anti-Runx2 was from Invitrogen, Carlsbad, CA, USA. The anti-PIP antibody (ab 62363) was purchased from abcam Inc., Cambridge, MA; and anti-GAPDH (V-18), anti-TGFß-RIII (Sc 28975), and anti-SDF-1 (Sc-28876) antibodies were purchased from Santa Cruz Biotechnology Inc, Santa Cruz, CA, USA. The mouse monoclonal anti-Myc and anti-VEGFA antibodies were developed and kindly provided by USC's Dr. Prakash S. Gill and Dr. Young Hong, respectively. The mouse monoclonal anti-Tubulin antibody, developed by Dr. Charles Walsh, was obtained from the Developmental Studies Hybridoma Bank under the auspices of the NICHD and The University of Iowa, Department of Biological Sciences, Iowa City, USA.

Plasmid construction

The cDNA encoding mouse Runx2 (MASN isoform, type-2), which is 97% identical to human type-2 Runx2 [1], was amplified using pcDNA3.0-Runx2 as template and the Flag epitope was inserted during the PCR amplification (see primers in additional file 8). The Flag-Runx2 cDNA was initially cloned into the SpeI/MfeI-digested lentiviral entry vector pEN_TmiRc3 (ATCC® catalog: MBA-248), and the resulting plasmid was recombined using Gateway® LR Clonase® II enzyme mix (Invitrogen, Carlsbad, CA, USA) with the pSLIK (single lentivector for inducible knockdown) destination vector carrying a hygromycin resistance gene (ATCC® catalog: MBA-237). The entry and destination vectors were kindly provided by USC's Dr. Elizabeth Lowler (Childrens Hospital Los Angeles). The DNA-binding mutant Runx2-M was constructed by site directed mutagenesis of two arginine residues at positions 265 and 268 (GRSGRGK) known to contact DNA in the crystal structure [40]. Lentiviral plasmid for constitutive Luciferase expression pCCL-c-MNDU3c-Luc-PGK-eGFP was kindly provided by USC's Dr. Michael Kahn at the Zilkha Neurogenetic Institute.

Lentivirus production and infection

For packaging, the lentiviral expression plasmids were cotransfected by the calcium chloride method into HEK293T cells along with helper plasmids pMD.G1 and pCMVR8.91[106, 107]. Culture media containing viral particles were harvested after 48-72 hours and used for transduction of C4-2B cells in the presence of 8 μg/ml Polybrene (Millipore Corp., MA, USA). After infection with the lentiviruses, the transduced cells were selected with 50 μg/ml of Hygromycin.

Transient transfection and Luciferase assays

Transient transfection and Luciferase assays were performed essentially as described by Khalid et al. [99]. Briefly, 25,000 cells were plated in a 24-well plate 48 hours prior to transfection using Invitrogen's Lipofectamine™ LTX reagent according to manufacturer's instructions. Cells were harvested and subjected to luciferase assay using Luciferase Assay system from Promega, Madison, WI, USA.

High throughput gene expression measurement and analysis

Gene expression profiling was performed using the BeadChip™ platform (Illumina) and chip reference 8, version 3 for humans, which contains 24,526 gene probes and 664 negative control probes. Details of the raw data processing and analysis are provided in the additional file 9. Briefly, after background correction, the normalized expression intensities for all probes were subjected to a two-way analysis of variance (ANOVA) and the resulting p-values were adjusted for multiple testing using the Benjimini-Hochberg method [108]. The differentially expressed probes were further investigated using the Ingenuity Pathways Analysis package (IPA™; http://​www.​ingenuity.​com) to identify biological functions and disease categories that are significantly enriched among the differentially expressed genes. Right-tailed Fisher's exact test as implemented in the IPA software was used to calculate a p-value for the probability of each network to be enriched for Runx2-regulated genes due to chance alone. The microarray data has been deposited to the GEO database with the accession code GSE24261.

Preparation of conditioned medium and gelatin zymography

C4-2B/Rx2dox cells were cultured in 10 cm culture dishes to 80% confluence, washed 3 times with RPMI-1640 and treated with Dox in 10 mL of RPMI-1640 without FBS for 24 hrs. For zymography the 25 μl of the conditioned media from Dox treated or control cells were analyzed by 8% acrylamide gels containing 0.1% w/v gelatin [109] After electrophoresis, the gel was washed with 2.5% Triton X-100 and incubated in the developing buffer (500 mM Tris-HCl, pH 7.8, 2 M NaCl, 50 mM CaCl2) overnight to induce gelatin lysis. Gel was stained by Coomasie blue-250. For western blot analysis of conditioned media, 250 μl of the supernatant was TCA-precipitated and subjected to reducing SDS-PAGE analysis using standard procedures.

In vitro invasion and migration assays

Invasion through Matrigel™ was assessed by incubating 20,000 luciferase-expressing cells at the top of 24 well BD BioCoat™ Growth Factor Reduced Matrigel™ chamber (BD Biosciences, MA, USA). Migration was assessed using BD BioCoat™ Control Inserts. Cells that migrated or invaded to the bottom compartment were visualized by Diff-Quick™ staining kit (Baxter, MN, USA) or subjected to luciferase assays for quantification. Invasion index was defined as the percentage of cells that invaded through Matrigel™ over those that migrated under the same conditions but without the Matrigel™.

Proliferation, cell cycle and apoptosis

Cultures on different days were subjected to MTT assay (Sigma, St Louis, MO) to measure viable cells in culture. For cell cycle analysis, 2 × 105 to 1 × 106 cells were harvested, washed twice with PBS (1 mL) at room temperature and stored in absolute ethanol (4 mL) for at least 24 hours. Pelleted cells were rehydrated in 5 mL PBS for 15 minutes, followed by staining with 1 mL of a propidium iodide (PI, Calibiochem, La jolla, CA, USA) solution containing 3 μM PI in incubation buffer (100 mM Tris, pH 7.4, 150 mM NaCl, 1 mM CaCl2, 0.5 mM MgCl2, 0.1% Nonidet® P-40). The cell suspension (1 mL) was subjected to fluorescence-activated cell sorting (FACScaliber, Becton Dickinson, MA, USA) and each cell was assigned to the G1, S, G2 or M phase of the cell cycle based on the PI intensity and using the Multicycle v3.0 software (Phoenix Flow Systems, San Diego, CA, USA). To assess apoptosis, cells were lysed in caspase assay buffer containing 50 mM HEPES (pH 7.5), 100 mM NaCl, 2 mM EDTA, 0.1% CHAPS, 10% sucrose, and 5 mM DTT. Aliquots of crude cell lysate (50 μg protein) were incubated with the caspase-3 substrate Ac-DEVD-AMC (EMD/Calbiochem, La Jolla, CA, USA) at 37°C for 30 min and the caspase-3 activity was quantified by flow fluorimetry with excitation at 380 nm and emission at 440 nm using Victor3V™ from PerkinElmer, Shelton, CT, USA.

Western blot analysis

Between 1 × 105 and 2 × 105 cells were washed once with PBS and lysed with 200 μL of incubation buffer [100 mM Tris (pH 7.4), 500 mM NaCl, 1 mM CaCl2, 0.5 mM MgCl2, 0.1% Nonidet® P-40] supplemented with Complete™ protease inhibitor mix (Roche Diagnostics, Indianapolis, IN, USA). Aliquots of 40 μg cell lysate were mixed with an equal volume of Laemmli buffer and proteins were resolved by 10% SDS-PAGE, transferred to Amersham Hybond™-P PVDF (GE Healthcare, Piscataway, NJ, USA) membranes, and visualized using respective antibodies and the Western Lightning™ Plus-ECL kit (PerkinElmer Inc, Waltham, MA, USA) followed by exposure to X-ray film (ISCBioExpress®, Kaysville, UT, USA).

RT-qPCR

Total RNA was isolated using Aurum Total RNA kit (Bio-Rad Laboratories Inc., Hercules, CA, USA) following the manufacturer's recommendations and 1 μg was reverse transcribed using the Superscript III kit (Invitrogen, CA, USA). The cDNA was subjected to real-time PCR amplification using iQ SYBR Green Supermix and a Opticon™2 real time PCR machine from Bio-Rad, Hercules, CA. The sequences of primers for amplification of the cDNA of interest and the control GAPDH are listed in the additional file 8.
GEO accession code for the microarray data: GSE24261

Acknowledgements

We thank Dr. Joseph Hacia and Dr. Nyam-Osor Chimge at the University of Southern California for their critical comments during the preparation of the manuscript. We also thank Dr. Gerard Karsenty (Columbia University, New York, NY) for the 6XOSE2-luciferase reporter and pCDNA3.1-Runx2 (mouse) plasmids. The microarray analysis was performed at the Southern California Genotyping Consortium at UCLA http://​scgc.​genetics.​ucla.​edu/​ under the direction of Mr. Joseph DeYoung. Further analysis of the gene expression profiles was performed with help from Dr. Yibu Chen at the USC Norris Medical Library. We thank Dr. Shinwu Jeong and Dr. Gabriel M Gordon for their help with the zymography assay. We also thank Dr. Pradip Roy-Burman and Helty Adisetiyo for kindly providing the PC3high cell line. The anti-TGFß-RIII antibody was kindly provided by Dr. Yang Chai at Center for Craniofacial Molecular Biology; and anti-SDF-1 antibody was kindly provided by Dr. Henry Sucov at the University of Southern California. This work was funded by NIH grants R01 CA 109147 and R01 CA 136924. SKB was supported by a postdoctoral Innovative Chapter Research Award; and YG by a Meyer Young Investigator Fellowship, both from the Arthritis Foundation Southern California Chapter. BF holds the J. Harold and Edna L. LaBriola Chair in Genetic Orthopedic Research at USC.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

Conceived and designed the experiments: SKB and BF. Performed the experiments: SKB, OK, YG, AEG, SS and DJP. Bioinformatics analysis: RRS and DM. Analyzed data and wrote the paper: SKB, GAC and BF. All authors have read and approved the final manuscript.
Anhänge

Electronic supplementary material

Literatur
1.
Zurück zum Zitat Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G: Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell. 1997, 89: 747-754. 10.1016/S0092-8674(00)80257-3PubMed Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G: Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell. 1997, 89: 747-754. 10.1016/S0092-8674(00)80257-3PubMed
2.
Zurück zum Zitat Cameron ER, Neil JC: The Runx genes: lineage-specific oncogenes and tumor suppressors. Oncogene. 2004, 23: 4308-4314. 10.1038/sj.onc.1207130PubMed Cameron ER, Neil JC: The Runx genes: lineage-specific oncogenes and tumor suppressors. Oncogene. 2004, 23: 4308-4314. 10.1038/sj.onc.1207130PubMed
3.
Zurück zum Zitat Lo Coco F, Pisegna S, Diverio D: The AML1 gene: a transcription factor involved in the pathogenesis of myeloid and lymphoid leukemias. Haematologica. 1997, 82: 364-370.PubMed Lo Coco F, Pisegna S, Diverio D: The AML1 gene: a transcription factor involved in the pathogenesis of myeloid and lymphoid leukemias. Haematologica. 1997, 82: 364-370.PubMed
4.
Zurück zum Zitat Woolf E, Xiao C, Fainaru O, Lotem J, Rosen D, Negreanu V, Bernstein Y, Goldenberg D, Brenner O, Berke G: Runx3 and Runx1 are required for CD8 T cell development during thymopoiesis. Proc Natl Acad Sci USA. 2003, 100: 7731-7736. 10.1073/pnas.1232420100PubMedCentralPubMed Woolf E, Xiao C, Fainaru O, Lotem J, Rosen D, Negreanu V, Bernstein Y, Goldenberg D, Brenner O, Berke G: Runx3 and Runx1 are required for CD8 T cell development during thymopoiesis. Proc Natl Acad Sci USA. 2003, 100: 7731-7736. 10.1073/pnas.1232420100PubMedCentralPubMed
5.
Zurück zum Zitat Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M: Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell. 1997, 89: 755-764. 10.1016/S0092-8674(00)80258-5PubMed Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M: Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell. 1997, 89: 755-764. 10.1016/S0092-8674(00)80258-5PubMed
6.
Zurück zum Zitat Otto F, Thornell AP, Crompton T, Denzel A, Gilmour KC, Rosewell IR, Stamp GW, Beddington RS, Mundlos S, Olsen BR: Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell. 1997, 89: 765-771. 10.1016/S0092-8674(00)80259-7PubMed Otto F, Thornell AP, Crompton T, Denzel A, Gilmour KC, Rosewell IR, Stamp GW, Beddington RS, Mundlos S, Olsen BR: Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell. 1997, 89: 765-771. 10.1016/S0092-8674(00)80259-7PubMed
7.
Zurück zum Zitat Levanon D, Bettoun D, Harris-Cerruti C, Woolf E, Negreanu V, Eilam R, Bernstein Y, Goldenberg D, Xiao C, Fliegauf M: The Runx3 transcription factor regulates development and survival of TrkC dorsal root ganglia neurons. Embo J. 2002, 21: 3454-3463. 10.1093/emboj/cdf370PubMedCentralPubMed Levanon D, Bettoun D, Harris-Cerruti C, Woolf E, Negreanu V, Eilam R, Bernstein Y, Goldenberg D, Xiao C, Fliegauf M: The Runx3 transcription factor regulates development and survival of TrkC dorsal root ganglia neurons. Embo J. 2002, 21: 3454-3463. 10.1093/emboj/cdf370PubMedCentralPubMed
8.
Zurück zum Zitat Ito Y: Oncogenic potential of the RUNX gene family: 'overview'. Oncogene. 2004, 23: 4198-4208. 10.1038/sj.onc.1207755PubMed Ito Y: Oncogenic potential of the RUNX gene family: 'overview'. Oncogene. 2004, 23: 4198-4208. 10.1038/sj.onc.1207755PubMed
9.
Zurück zum Zitat Bae SC, Choi JK: Tumor suppressor activity of RUNX3. Oncogene. 2004, 23: 4336-4340. 10.1038/sj.onc.1207286PubMed Bae SC, Choi JK: Tumor suppressor activity of RUNX3. Oncogene. 2004, 23: 4336-4340. 10.1038/sj.onc.1207286PubMed
10.
Zurück zum Zitat Levanon D, Groner Y: Structure and regulated expression of mammalian RUNX genes. Oncogene. 2004, 23: 4211-4219. 10.1038/sj.onc.1207670PubMed Levanon D, Groner Y: Structure and regulated expression of mammalian RUNX genes. Oncogene. 2004, 23: 4211-4219. 10.1038/sj.onc.1207670PubMed
11.
Zurück zum Zitat Oshimo Y, Oue N, Mitani Y, Nakayama H, Kitadai Y, Yoshida K, Ito Y, Chayama K, Yasui W: Frequent loss of RUNX3 expression by promoter hypermethylation in gastric carcinoma. Pathobiology. 2004, 71: 137-143. 10.1159/000076468PubMed Oshimo Y, Oue N, Mitani Y, Nakayama H, Kitadai Y, Yoshida K, Ito Y, Chayama K, Yasui W: Frequent loss of RUNX3 expression by promoter hypermethylation in gastric carcinoma. Pathobiology. 2004, 71: 137-143. 10.1159/000076468PubMed
12.
Zurück zum Zitat Rossetti S, Van Unen L, Touw IP, Hoogeveen AT, Sacchi N: Myeloid maturation block by AML1-MTG16 is associated with Csf1r epigenetic downregulation. Oncogene. 2005, 24: 5325-5332. 10.1038/sj.onc.1208651PubMed Rossetti S, Van Unen L, Touw IP, Hoogeveen AT, Sacchi N: Myeloid maturation block by AML1-MTG16 is associated with Csf1r epigenetic downregulation. Oncogene. 2005, 24: 5325-5332. 10.1038/sj.onc.1208651PubMed
13.
Zurück zum Zitat Kilbey A, Blyth K, Wotton S, Terry A, Jenkins A, Bell M, Hanlon L, Cameron ER, Neil JC: Runx2 disruption promotes immortalization and confers resistance to oncogene-induced senescence in primary murine fibroblasts. Cancer Res. 2007, 67: 11263-11271. 10.1158/0008-5472.CAN-07-3016PubMedCentralPubMed Kilbey A, Blyth K, Wotton S, Terry A, Jenkins A, Bell M, Hanlon L, Cameron ER, Neil JC: Runx2 disruption promotes immortalization and confers resistance to oncogene-induced senescence in primary murine fibroblasts. Cancer Res. 2007, 67: 11263-11271. 10.1158/0008-5472.CAN-07-3016PubMedCentralPubMed
14.
Zurück zum Zitat Zaidi SK, Pande S, Pratap J, Gaur T, Grigoriu S, Ali SA, Stein JL, Lian JB, van Wijnen AJ, Stein GS: Runx2 deficiency and defective subnuclear targeting bypass senescence to promote immortalization and tumorigenic potential. Proc Natl Acad Sci USA. 2007, 104: 19861-19866. 10.1073/pnas.0709650104PubMedCentralPubMed Zaidi SK, Pande S, Pratap J, Gaur T, Grigoriu S, Ali SA, Stein JL, Lian JB, van Wijnen AJ, Stein GS: Runx2 deficiency and defective subnuclear targeting bypass senescence to promote immortalization and tumorigenic potential. Proc Natl Acad Sci USA. 2007, 104: 19861-19866. 10.1073/pnas.0709650104PubMedCentralPubMed
15.
Zurück zum Zitat Galindo M, Pratap J, Young DW, Hovhannisyan H, Im HJ, Choi JY, Lian JB, Stein JL, Stein GS, van Wijnen AJ: The bone-specific expression of Runx2 oscillates during the cell cycle to support a G1-related antiproliferative function in osteoblasts. J Biol Chem. 2005, 280: 20274-20285. 10.1074/jbc.M413665200PubMedCentralPubMed Galindo M, Pratap J, Young DW, Hovhannisyan H, Im HJ, Choi JY, Lian JB, Stein JL, Stein GS, van Wijnen AJ: The bone-specific expression of Runx2 oscillates during the cell cycle to support a G1-related antiproliferative function in osteoblasts. J Biol Chem. 2005, 280: 20274-20285. 10.1074/jbc.M413665200PubMedCentralPubMed
16.
Zurück zum Zitat Pratap J, Galindo M, Zaidi SK, Vradii D, Bhat BM, Robinson JA, Choi JY, Komori T, Stein JL, Lian JB: Cell growth regulatory role of Runx2 during proliferative expansion of preosteoblasts. Cancer Res. 2003, 63: 5357-5362.PubMed Pratap J, Galindo M, Zaidi SK, Vradii D, Bhat BM, Robinson JA, Choi JY, Komori T, Stein JL, Lian JB: Cell growth regulatory role of Runx2 during proliferative expansion of preosteoblasts. Cancer Res. 2003, 63: 5357-5362.PubMed
17.
Zurück zum Zitat Thomas DM, Johnson SA, Sims NA, Trivett MK, Slavin JL, Rubin BP, Waring P, McArthur GA, Walkley CR, Holloway AJ: Terminal osteoblast differentiation, mediated by runx2 and p27KIP1, is disrupted in osteosarcoma. J Cell Biol. 2004, 167: 925-934. 10.1083/jcb.200409187PubMedCentralPubMed Thomas DM, Johnson SA, Sims NA, Trivett MK, Slavin JL, Rubin BP, Waring P, McArthur GA, Walkley CR, Holloway AJ: Terminal osteoblast differentiation, mediated by runx2 and p27KIP1, is disrupted in osteosarcoma. J Cell Biol. 2004, 167: 925-934. 10.1083/jcb.200409187PubMedCentralPubMed
18.
Zurück zum Zitat Blyth K, Terry A, Mackay N, Vaillant F, Bell M, Cameron ER, Neil JC, Stewart M: Runx2: a novel oncogenic effector revealed by in vivo complementation and retroviral tagging. Oncogene. 2001, 20: 295-302. 10.1038/sj.onc.1204090PubMed Blyth K, Terry A, Mackay N, Vaillant F, Bell M, Cameron ER, Neil JC, Stewart M: Runx2: a novel oncogenic effector revealed by in vivo complementation and retroviral tagging. Oncogene. 2001, 20: 295-302. 10.1038/sj.onc.1204090PubMed
19.
Zurück zum Zitat Blyth K, Cameron ER, Neil JC: The RUNX genes: gain or loss of function in cancer. Nat Rev Cancer. 2005, 5: 376-387. 10.1038/nrc1607PubMed Blyth K, Cameron ER, Neil JC: The RUNX genes: gain or loss of function in cancer. Nat Rev Cancer. 2005, 5: 376-387. 10.1038/nrc1607PubMed
20.
Zurück zum Zitat Komori T: Regulation of osteoblast differentiation by transcription factors. J Cell Biochem. 2006, 99: 1233-1239. 10.1002/jcb.20958PubMed Komori T: Regulation of osteoblast differentiation by transcription factors. J Cell Biochem. 2006, 99: 1233-1239. 10.1002/jcb.20958PubMed
21.
Zurück zum Zitat Otto F, Kanegane H, Mundlos S: Mutations in the RUNX2 gene in patients with cleidocranial dysplasia. Hum Mutat. 2002, 19: 209-216. 10.1002/humu.10043PubMed Otto F, Kanegane H, Mundlos S: Mutations in the RUNX2 gene in patients with cleidocranial dysplasia. Hum Mutat. 2002, 19: 209-216. 10.1002/humu.10043PubMed
22.
Zurück zum Zitat Akech J, Wixted JJ, Bedard K, van der Deen M, Hussain S, Guise TA, van Wijnen AJ, Stein JL, Languino LR, Altieri DC: Runx2 association with progression of prostate cancer in patients: mechanisms mediating bone osteolysis and osteoblastic metastatic lesions. Oncogene. 2009, 811-821. Akech J, Wixted JJ, Bedard K, van der Deen M, Hussain S, Guise TA, van Wijnen AJ, Stein JL, Languino LR, Altieri DC: Runx2 association with progression of prostate cancer in patients: mechanisms mediating bone osteolysis and osteoblastic metastatic lesions. Oncogene. 2009, 811-821.
23.
Zurück zum Zitat Koeneman KS, Yeung F, Chung LW: Osteomimetic properties of prostate cancer cells: a hypothesis supporting the predilection of prostate cancer metastasis and growth in the bone environment. Prostate. 1999, 39: 246-261. 10.1002/(SICI)1097-0045(19990601)39:4<246::AID-PROS5>3.0.CO;2-UPubMed Koeneman KS, Yeung F, Chung LW: Osteomimetic properties of prostate cancer cells: a hypothesis supporting the predilection of prostate cancer metastasis and growth in the bone environment. Prostate. 1999, 39: 246-261. 10.1002/(SICI)1097-0045(19990601)39:4<246::AID-PROS5>3.0.CO;2-UPubMed
24.
Zurück zum Zitat Chua CW, Chiu YT, Yuen HF, Chan KW, Man K, Wang X, Ling MT, Wong YC: Suppression of androgen-independent prostate cancer cell aggressiveness by FTY720: validating Runx2 as a potential antimetastatic drug screening platform. Clin Cancer Res. 2009, 15: 4322-4335. 10.1158/1078-0432.CCR-08-3157PubMed Chua CW, Chiu YT, Yuen HF, Chan KW, Man K, Wang X, Ling MT, Wong YC: Suppression of androgen-independent prostate cancer cell aggressiveness by FTY720: validating Runx2 as a potential antimetastatic drug screening platform. Clin Cancer Res. 2009, 15: 4322-4335. 10.1158/1078-0432.CCR-08-3157PubMed
25.
Zurück zum Zitat Lin DL, Tarnowski CP, Zhang J, Dai J, Rohn E, Patel AH, Morris MD, Keller ET: Bone metastatic LNCaP-derivative C4-2B prostate cancer cell line mineralizes in vitro. Prostate. 2001, 47: 212-221. 10.1002/pros.1065PubMed Lin DL, Tarnowski CP, Zhang J, Dai J, Rohn E, Patel AH, Morris MD, Keller ET: Bone metastatic LNCaP-derivative C4-2B prostate cancer cell line mineralizes in vitro. Prostate. 2001, 47: 212-221. 10.1002/pros.1065PubMed
26.
Zurück zum Zitat Selvamurugan N, Kwok S, Partridge NC: Smad3 interacts with JunB and Cbfa1/Runx2 for transforming growth factor-beta1-stimulated collagenase-3 expression in human breast cancer cells. J Biol Chem. 2004, 279: 27764-27773. 10.1074/jbc.M312870200PubMed Selvamurugan N, Kwok S, Partridge NC: Smad3 interacts with JunB and Cbfa1/Runx2 for transforming growth factor-beta1-stimulated collagenase-3 expression in human breast cancer cells. J Biol Chem. 2004, 279: 27764-27773. 10.1074/jbc.M312870200PubMed
27.
Zurück zum Zitat Lim M, Zhong C, Yang S, Bell AM, Cohen MB, Roy-Burman P: Runx2 regulates survivin expression in prostate cancer cells. Lab Invest. 90: 222-233. Lim M, Zhong C, Yang S, Bell AM, Cohen MB, Roy-Burman P: Runx2 regulates survivin expression in prostate cancer cells. Lab Invest. 90: 222-233.
28.
Zurück zum Zitat Pratap J, Javed A, Languino LR, van Wijnen AJ, Stein JL, Stein GS, Lian JB: The Runx2 osteogenic transcription factor regulates matrix metalloproteinase 9 in bone metastatic cancer cells and controls cell invasion. Mol Cell Biol. 2005, 25: 8581-8591. 10.1128/MCB.25.19.8581-8591.2005PubMedCentralPubMed Pratap J, Javed A, Languino LR, van Wijnen AJ, Stein JL, Stein GS, Lian JB: The Runx2 osteogenic transcription factor regulates matrix metalloproteinase 9 in bone metastatic cancer cells and controls cell invasion. Mol Cell Biol. 2005, 25: 8581-8591. 10.1128/MCB.25.19.8581-8591.2005PubMedCentralPubMed
29.
Zurück zum Zitat Barnes GL, Javed A, Waller SM, Kamal MH, Hebert KE, Hassan MQ, Bellahcene A, Van Wijnen AJ, Young MF, Lian JB: Osteoblast-related transcription factors Runx2 (Cbfa1/AML3) and MSX2 mediate the expression of bone sialoprotein in human metastatic breast cancer cells. Cancer Res. 2003, 63: 2631-2637.PubMed Barnes GL, Javed A, Waller SM, Kamal MH, Hebert KE, Hassan MQ, Bellahcene A, Van Wijnen AJ, Young MF, Lian JB: Osteoblast-related transcription factors Runx2 (Cbfa1/AML3) and MSX2 mediate the expression of bone sialoprotein in human metastatic breast cancer cells. Cancer Res. 2003, 63: 2631-2637.PubMed
30.
Zurück zum Zitat Zelzer E, Glotzer DJ, Hartmann C, Thomas D, Fukai N, Soker S, Olsen BR: Tissue specific regulation of VEGF expression during bone development requires Cbfa1/Runx2. Mech Dev. 2001, 106: 97-106. 10.1016/S0925-4773(01)00428-2PubMed Zelzer E, Glotzer DJ, Hartmann C, Thomas D, Fukai N, Soker S, Olsen BR: Tissue specific regulation of VEGF expression during bone development requires Cbfa1/Runx2. Mech Dev. 2001, 106: 97-106. 10.1016/S0925-4773(01)00428-2PubMed
31.
Zurück zum Zitat Geoffroy V, Kneissel M, Fournier B, Boyde A, Matthias P: High bone resorption in adult aging transgenic mice overexpressing cbfa1/runx2 in cells of the osteoblastic lineage. Mol Cell Biol. 2002, 22: 6222-6233. 10.1128/MCB.22.17.6222-6233.2002PubMedCentralPubMed Geoffroy V, Kneissel M, Fournier B, Boyde A, Matthias P: High bone resorption in adult aging transgenic mice overexpressing cbfa1/runx2 in cells of the osteoblastic lineage. Mol Cell Biol. 2002, 22: 6222-6233. 10.1128/MCB.22.17.6222-6233.2002PubMedCentralPubMed
32.
Zurück zum Zitat Maruyama Z, Yoshida CA, Furuichi T, Amizuka N, Ito M, Fukuyama R, Miyazaki T, Kitaura H, Nakamura K, Fujita T: Runx2 determines bone maturity and turnover rate in postnatal bone development and is involved in bone loss in estrogen deficiency. Dev Dyn. 2007, 236: 1876-1890. 10.1002/dvdy.21187PubMed Maruyama Z, Yoshida CA, Furuichi T, Amizuka N, Ito M, Fukuyama R, Miyazaki T, Kitaura H, Nakamura K, Fujita T: Runx2 determines bone maturity and turnover rate in postnatal bone development and is involved in bone loss in estrogen deficiency. Dev Dyn. 2007, 236: 1876-1890. 10.1002/dvdy.21187PubMed
33.
Zurück zum Zitat Yeung F, Law WK, Yeh CH, Westendorf JJ, Zhang Y, Wang R, Kao C, Chung LW: Regulation of human osteocalcin promoter in hormone-independent human prostate cancer cells. J Biol Chem. 2002, 277: 2468-2476. 10.1074/jbc.M105947200PubMed Yeung F, Law WK, Yeh CH, Westendorf JJ, Zhang Y, Wang R, Kao C, Chung LW: Regulation of human osteocalcin promoter in hormone-independent human prostate cancer cells. J Biol Chem. 2002, 277: 2468-2476. 10.1074/jbc.M105947200PubMed
34.
Zurück zum Zitat Pratap J, Imbalzano KM, Underwood JM, Cohet N, Gokul K, Akech J, van Wijnen AJ, Stein JL, Imbalzano AN, Nickerson JA: Ectopic runx2 expression in mammary epithelial cells disrupts formation of normal acini structure: implications for breast cancer progression. Cancer Res. 2009, 69: 6807-6814. 10.1158/0008-5472.CAN-09-1471PubMedCentralPubMed Pratap J, Imbalzano KM, Underwood JM, Cohet N, Gokul K, Akech J, van Wijnen AJ, Stein JL, Imbalzano AN, Nickerson JA: Ectopic runx2 expression in mammary epithelial cells disrupts formation of normal acini structure: implications for breast cancer progression. Cancer Res. 2009, 69: 6807-6814. 10.1158/0008-5472.CAN-09-1471PubMedCentralPubMed
35.
Zurück zum Zitat Kayed H, Jiang X, Keleg S, Jesnowski R, Giese T, Berger MR, Esposito I, Lohr M, Friess H, Kleeff J: Regulation and functional role of the Runt-related transcription factor-2 in pancreatic cancer. Br J Cancer. 2007, 97: 1106-1115. 10.1038/sj.bjc.6603984PubMedCentralPubMed Kayed H, Jiang X, Keleg S, Jesnowski R, Giese T, Berger MR, Esposito I, Lohr M, Friess H, Kleeff J: Regulation and functional role of the Runt-related transcription factor-2 in pancreatic cancer. Br J Cancer. 2007, 97: 1106-1115. 10.1038/sj.bjc.6603984PubMedCentralPubMed
36.
Zurück zum Zitat Endo T, Ohta K, Kobayashi T: Expression and function of Cbfa-1/Runx2 in thyroid papillary carcinoma cells. J Clin Endocrinol Metab. 2008, 93: 2409-2412. 10.1210/jc.2007-2805PubMed Endo T, Ohta K, Kobayashi T: Expression and function of Cbfa-1/Runx2 in thyroid papillary carcinoma cells. J Clin Endocrinol Metab. 2008, 93: 2409-2412. 10.1210/jc.2007-2805PubMed
37.
Zurück zum Zitat Horoszewicz JS, Leong SS, Kawinski E, Karr JP, Rosenthal H, Chu TM, Mirand EA, Murphy GP: LNCaP model of human prostatic carcinoma. Cancer Res. 1983, 43: 1809-1818.PubMed Horoszewicz JS, Leong SS, Kawinski E, Karr JP, Rosenthal H, Chu TM, Mirand EA, Murphy GP: LNCaP model of human prostatic carcinoma. Cancer Res. 1983, 43: 1809-1818.PubMed
38.
Zurück zum Zitat Wu HC, Hsieh JT, Gleave ME, Brown NM, Pathak S, Chung LW: Derivation of androgen-independent human LNCaP prostatic cancer cell sublines: role of bone stromal cells. Int J Cancer. 1994, 57: 406-412. 10.1002/ijc.2910570319PubMed Wu HC, Hsieh JT, Gleave ME, Brown NM, Pathak S, Chung LW: Derivation of androgen-independent human LNCaP prostatic cancer cell sublines: role of bone stromal cells. Int J Cancer. 1994, 57: 406-412. 10.1002/ijc.2910570319PubMed
39.
Zurück zum Zitat Shin KJ, Wall EA, Zavzavadjian JR, Santat LA, Liu J, Hwang JI, Rebres R, Roach T, Seaman W, Simon MI, Fraser ID: A single lentiviral vector platform for microRNA-based conditional RNA interference and coordinated transgene expression. Proc Natl Acad Sci USA. 2006, 103: 13759-13764. 10.1073/pnas.0606179103PubMedCentralPubMed Shin KJ, Wall EA, Zavzavadjian JR, Santat LA, Liu J, Hwang JI, Rebres R, Roach T, Seaman W, Simon MI, Fraser ID: A single lentiviral vector platform for microRNA-based conditional RNA interference and coordinated transgene expression. Proc Natl Acad Sci USA. 2006, 103: 13759-13764. 10.1073/pnas.0606179103PubMedCentralPubMed
40.
Zurück zum Zitat Tahirov TH, Inoue-Bungo T, Morii H, Fujikawa A, Sasaki M, Kimura K, Shiina M, Sato K, Kumasaka T, Yamamoto M: Structural analyses of DNA recognition by the AML1/Runx-1 Runt domain and its allosteric control by CBFbeta. Cell. 2001, 104: 755-767. 10.1016/S0092-8674(01)00271-9PubMed Tahirov TH, Inoue-Bungo T, Morii H, Fujikawa A, Sasaki M, Kimura K, Shiina M, Sato K, Kumasaka T, Yamamoto M: Structural analyses of DNA recognition by the AML1/Runx-1 Runt domain and its allosteric control by CBFbeta. Cell. 2001, 104: 755-767. 10.1016/S0092-8674(01)00271-9PubMed
41.
Zurück zum Zitat Sudhakar S, Li Y, Katz MS, Elango N: Translational regulation is a control point in RUNX2/Cbfa1 gene expression. Biochem Biophys Res Commun. 2001, 289: 616-622. 10.1006/bbrc.2001.6033PubMed Sudhakar S, Li Y, Katz MS, Elango N: Translational regulation is a control point in RUNX2/Cbfa1 gene expression. Biochem Biophys Res Commun. 2001, 289: 616-622. 10.1006/bbrc.2001.6033PubMed
42.
Zurück zum Zitat Ducy P, Karsenty G: Two distinct osteoblast-specific cis-acting elements control expression of a mouse osteocalcin gene. Mol Cell Biol. 1995, 15: 1858-1869.PubMedCentralPubMed Ducy P, Karsenty G: Two distinct osteoblast-specific cis-acting elements control expression of a mouse osteocalcin gene. Mol Cell Biol. 1995, 15: 1858-1869.PubMedCentralPubMed
43.
Zurück zum Zitat Vaes BL, Ducy P, Sijbers AM, Hendriks JM, van Someren EP, de Jong NG, van den Heuvel ER, Olijve W, van Zoelen EJ, Dechering KJ: Microarray analysis on Runx2-deficient mouse embryos reveals novel Runx2 functions and target genes during intramembranous and endochondral bone formation. Bone. 2006, 39: 724-738. 10.1016/j.bone.2006.04.024PubMed Vaes BL, Ducy P, Sijbers AM, Hendriks JM, van Someren EP, de Jong NG, van den Heuvel ER, Olijve W, van Zoelen EJ, Dechering KJ: Microarray analysis on Runx2-deficient mouse embryos reveals novel Runx2 functions and target genes during intramembranous and endochondral bone formation. Bone. 2006, 39: 724-738. 10.1016/j.bone.2006.04.024PubMed
44.
Zurück zum Zitat Cheung M, Chaboissier MC, Mynett A, Hirst E, Schedl A, Briscoe J: The transcriptional control of trunk neural crest induction, survival, and delamination. Dev Cell. 2005, 8: 179-192. 10.1016/j.devcel.2004.12.010PubMed Cheung M, Chaboissier MC, Mynett A, Hirst E, Schedl A, Briscoe J: The transcriptional control of trunk neural crest induction, survival, and delamination. Dev Cell. 2005, 8: 179-192. 10.1016/j.devcel.2004.12.010PubMed
45.
Zurück zum Zitat Yang J, Bielenberg DR, Rodig SJ, Doiron R, Clifton MC, Kung AL, Strong RK, Zurakowski D, Moses MA: Lipocalin 2 promotes breast cancer progression. Proc Natl Acad Sci USA. 2009, 106: 3913-3918. 10.1073/pnas.0810617106PubMedCentralPubMed Yang J, Bielenberg DR, Rodig SJ, Doiron R, Clifton MC, Kung AL, Strong RK, Zurakowski D, Moses MA: Lipocalin 2 promotes breast cancer progression. Proc Natl Acad Sci USA. 2009, 106: 3913-3918. 10.1073/pnas.0810617106PubMedCentralPubMed
46.
Zurück zum Zitat Caputo E, Manco G, Mandrich L, Guardiola J: A novel aspartyl proteinase from apocrine epithelia and breast tumors. J Biol Chem. 2000, 275: 7935-7941. 10.1074/jbc.275.11.7935PubMed Caputo E, Manco G, Mandrich L, Guardiola J: A novel aspartyl proteinase from apocrine epithelia and breast tumors. J Biol Chem. 2000, 275: 7935-7941. 10.1074/jbc.275.11.7935PubMed
47.
Zurück zum Zitat Chiang L, Contreras L, Chiang J, Ward PH: Human prostatic gastricsinogen: the precursor of seminal fluid acid proteinase. Arch Biochem Biophys. 1981, 210: 14-20. 10.1016/0003-9861(81)90158-2PubMed Chiang L, Contreras L, Chiang J, Ward PH: Human prostatic gastricsinogen: the precursor of seminal fluid acid proteinase. Arch Biochem Biophys. 1981, 210: 14-20. 10.1016/0003-9861(81)90158-2PubMed
48.
Zurück zum Zitat Morita M, Yoshiuchi N, Arakawa H, Nishimura S: CMAP: a novel cystatin-like gene involved in liver metastasis. Cancer Res. 1999, 59: 151-158.PubMed Morita M, Yoshiuchi N, Arakawa H, Nishimura S: CMAP: a novel cystatin-like gene involved in liver metastasis. Cancer Res. 1999, 59: 151-158.PubMed
49.
Zurück zum Zitat Sherbet GV: Metastasis promoter S100A4 is a potentially valuable molecular target for cancer therapy. Cancer Lett. 2009, 280: 15-30. 10.1016/j.canlet.2008.10.037PubMed Sherbet GV: Metastasis promoter S100A4 is a potentially valuable molecular target for cancer therapy. Cancer Lett. 2009, 280: 15-30. 10.1016/j.canlet.2008.10.037PubMed
50.
Zurück zum Zitat Matsuura I, Lai CY, Chiang KN: Functional interaction between Smad3 and S100A4 (metastatin-1) for TGF-beta-mediated cancer cell invasiveness. Biochem J. 2010, 426: 327-335. 10.1042/BJ20090990PubMed Matsuura I, Lai CY, Chiang KN: Functional interaction between Smad3 and S100A4 (metastatin-1) for TGF-beta-mediated cancer cell invasiveness. Biochem J. 2010, 426: 327-335. 10.1042/BJ20090990PubMed
51.
Zurück zum Zitat Artym VV, Zhang Y, Seillier-Moiseiwitsch F, Yamada KM, Mueller SC: Dynamic interactions of cortactin and membrane type 1 matrix metalloproteinase at invadopodia: defining the stages of invadopodia formation and function. Cancer Res. 2006, 66: 3034-3043. 10.1158/0008-5472.CAN-05-2177PubMed Artym VV, Zhang Y, Seillier-Moiseiwitsch F, Yamada KM, Mueller SC: Dynamic interactions of cortactin and membrane type 1 matrix metalloproteinase at invadopodia: defining the stages of invadopodia formation and function. Cancer Res. 2006, 66: 3034-3043. 10.1158/0008-5472.CAN-05-2177PubMed
52.
Zurück zum Zitat Larsen M, Artym VV, Green JA, Yamada KM: The matrix reorganized: extracellular matrix remodeling and integrin signaling. Curr Opin Cell Biol. 2006, 18: 463-471. 10.1016/j.ceb.2006.08.009PubMed Larsen M, Artym VV, Green JA, Yamada KM: The matrix reorganized: extracellular matrix remodeling and integrin signaling. Curr Opin Cell Biol. 2006, 18: 463-471. 10.1016/j.ceb.2006.08.009PubMed
53.
Zurück zum Zitat Schmidt KL, Marcus-Gueret N, Adeleye A, Webber J, Baillie D, Stringham EG: The cell migration molecule UNC-53/NAV2 is linked to the ARP2/3 complex by ABI-1. Development. 2009, 136: 563-574. 10.1242/dev.016816PubMed Schmidt KL, Marcus-Gueret N, Adeleye A, Webber J, Baillie D, Stringham EG: The cell migration molecule UNC-53/NAV2 is linked to the ARP2/3 complex by ABI-1. Development. 2009, 136: 563-574. 10.1242/dev.016816PubMed
54.
Zurück zum Zitat Sekerkova G, Loomis PA, Changyaleket B, Zheng L, Eytan R, Chen B, Mugnaini E, Bartles JR: Novel espin actin-bundling proteins are localized to Purkinje cell dendritic spines and bind the Src homology 3 adapter protein insulin receptor substrate p53. J Neurosci. 2003, 23: 1310-1319.PubMedCentralPubMed Sekerkova G, Loomis PA, Changyaleket B, Zheng L, Eytan R, Chen B, Mugnaini E, Bartles JR: Novel espin actin-bundling proteins are localized to Purkinje cell dendritic spines and bind the Src homology 3 adapter protein insulin receptor substrate p53. J Neurosci. 2003, 23: 1310-1319.PubMedCentralPubMed
55.
Zurück zum Zitat Riederer BM: Microtubule-associated protein 1B, a growth-associated and phosphorylated scaffold protein. Brain Res Bull. 2007, 71: 541-558. 10.1016/j.brainresbull.2006.11.012PubMed Riederer BM: Microtubule-associated protein 1B, a growth-associated and phosphorylated scaffold protein. Brain Res Bull. 2007, 71: 541-558. 10.1016/j.brainresbull.2006.11.012PubMed
56.
Zurück zum Zitat Maul RS, Song Y, Amann KJ, Gerbin SC, Pollard TD, Chang DD: EPLIN regulates actin dynamics by cross-linking and stabilizing filaments. J Cell Biol. 2003, 160: 399-407. 10.1083/jcb.200212057PubMedCentralPubMed Maul RS, Song Y, Amann KJ, Gerbin SC, Pollard TD, Chang DD: EPLIN regulates actin dynamics by cross-linking and stabilizing filaments. J Cell Biol. 2003, 160: 399-407. 10.1083/jcb.200212057PubMedCentralPubMed
57.
Zurück zum Zitat Paavilainen VO, Bertling E, Falck S, Lappalainen P: Regulation of cytoskeletal dynamics by actin-monomer-binding proteins. Trends Cell Biol. 2004, 14: 386-394. 10.1016/j.tcb.2004.05.002PubMed Paavilainen VO, Bertling E, Falck S, Lappalainen P: Regulation of cytoskeletal dynamics by actin-monomer-binding proteins. Trends Cell Biol. 2004, 14: 386-394. 10.1016/j.tcb.2004.05.002PubMed
58.
Zurück zum Zitat Levoye A, Balabanian K, Baleux F, Bachelerie F, Lagane B: CXCR7 heterodimerizes with CXCR4 and regulates CXCL12-mediated G protein signaling. Blood. 2009, 113: 6085-6093. 10.1182/blood-2008-12-196618PubMed Levoye A, Balabanian K, Baleux F, Bachelerie F, Lagane B: CXCR7 heterodimerizes with CXCR4 and regulates CXCL12-mediated G protein signaling. Blood. 2009, 113: 6085-6093. 10.1182/blood-2008-12-196618PubMed
59.
Zurück zum Zitat Mazzinghi B, Ronconi E, Lazzeri E, Sagrinati C, Ballerini L, Angelotti ML, Parente E, Mancina R, Netti GS, Becherucci F: Essential but differential role for CXCR4 and CXCR7 in the therapeutic homing of human renal progenitor cells. J Exp Med. 2008, 205: 479-490. 10.1084/jem.20071903PubMedCentralPubMed Mazzinghi B, Ronconi E, Lazzeri E, Sagrinati C, Ballerini L, Angelotti ML, Parente E, Mancina R, Netti GS, Becherucci F: Essential but differential role for CXCR4 and CXCR7 in the therapeutic homing of human renal progenitor cells. J Exp Med. 2008, 205: 479-490. 10.1084/jem.20071903PubMedCentralPubMed
60.
Zurück zum Zitat Kryczek I, Wei S, Keller E, Liu R, Zou W: Stroma-derived factor (SDF-1/CXCL12) and human tumor pathogenesis. Am J Physiol Cell Physiol. 2007, 292: C987-995. 10.1152/ajpcell.00406.2006PubMed Kryczek I, Wei S, Keller E, Liu R, Zou W: Stroma-derived factor (SDF-1/CXCL12) and human tumor pathogenesis. Am J Physiol Cell Physiol. 2007, 292: C987-995. 10.1152/ajpcell.00406.2006PubMed
61.
Zurück zum Zitat Singh S, Singh UP, Grizzle WE, Lillard JW: CXCL12-CXCR4 interactions modulate prostate cancer cell migration, metalloproteinase expression and invasion. Lab Invest. 2004, 84: 1666-1676. 10.1038/labinvest.3700181PubMed Singh S, Singh UP, Grizzle WE, Lillard JW: CXCL12-CXCR4 interactions modulate prostate cancer cell migration, metalloproteinase expression and invasion. Lab Invest. 2004, 84: 1666-1676. 10.1038/labinvest.3700181PubMed
62.
Zurück zum Zitat Kryczek I, Lange A, Mottram P, Alvarez X, Cheng P, Hogan M, Moons L, Wei S, Zou L, Machelon V: CXCL12 and vascular endothelial growth factor synergistically induce neoangiogenesis in human ovarian cancers. Cancer Res. 2005, 65: 465-472.PubMed Kryczek I, Lange A, Mottram P, Alvarez X, Cheng P, Hogan M, Moons L, Wei S, Zou L, Machelon V: CXCL12 and vascular endothelial growth factor synergistically induce neoangiogenesis in human ovarian cancers. Cancer Res. 2005, 65: 465-472.PubMed
63.
Zurück zum Zitat Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS, McCauley LK: Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res. 2002, 62: 1832-1837.PubMed Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS, McCauley LK: Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res. 2002, 62: 1832-1837.PubMed
64.
Zurück zum Zitat Ara T, Tokoyoda K, Sugiyama T, Egawa T, Kawabata K, Nagasawa T: Long-term hematopoietic stem cells require stromal cell-derived factor-1 for colonizing bone marrow during ontogeny. Immunity. 2003, 19: 257-267. 10.1016/S1074-7613(03)00201-2PubMed Ara T, Tokoyoda K, Sugiyama T, Egawa T, Kawabata K, Nagasawa T: Long-term hematopoietic stem cells require stromal cell-derived factor-1 for colonizing bone marrow during ontogeny. Immunity. 2003, 19: 257-267. 10.1016/S1074-7613(03)00201-2PubMed
65.
Zurück zum Zitat Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, Nagler A, Ben-Hur H, Many A, Shultz L: Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999, 283: 845-848. 10.1126/science.283.5403.845PubMed Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, Nagler A, Ben-Hur H, Many A, Shultz L: Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999, 283: 845-848. 10.1126/science.283.5403.845PubMed
66.
Zurück zum Zitat Waltregny D, Bellahcene A, Van Riet I, Fisher LW, Young M, Fernandez P, Dewe W, de Leval J, Castronovo V: Prognostic value of bone sialoprotein expression in clinically localized human prostate cancer. J Natl Cancer Inst. 1998, 90: 1000-1008. 10.1093/jnci/90.13.1000PubMed Waltregny D, Bellahcene A, Van Riet I, Fisher LW, Young M, Fernandez P, Dewe W, de Leval J, Castronovo V: Prognostic value of bone sialoprotein expression in clinically localized human prostate cancer. J Natl Cancer Inst. 1998, 90: 1000-1008. 10.1093/jnci/90.13.1000PubMed
67.
Zurück zum Zitat Waltregny D, Bellahcene A, de Leval X, Florkin B, Weidle U, Castronovo V: Increased expression of bone sialoprotein in bone metastases compared with visceral metastases in human breast and prostate cancers. J Bone Miner Res. 2000, 15: 834-843. 10.1359/jbmr.2000.15.5.834PubMed Waltregny D, Bellahcene A, de Leval X, Florkin B, Weidle U, Castronovo V: Increased expression of bone sialoprotein in bone metastases compared with visceral metastases in human breast and prostate cancers. J Bone Miner Res. 2000, 15: 834-843. 10.1359/jbmr.2000.15.5.834PubMed
68.
Zurück zum Zitat Zhang JH, Tang J, Wang J, Ma W, Zheng W, Yoneda T, Chen J: Over-expression of bone sialoprotein enhances bone metastasis of human breast cancer cells in a mouse model. Int J Oncol. 2003, 23: 1043-1048.PubMed Zhang JH, Tang J, Wang J, Ma W, Zheng W, Yoneda T, Chen J: Over-expression of bone sialoprotein enhances bone metastasis of human breast cancer cells in a mouse model. Int J Oncol. 2003, 23: 1043-1048.PubMed
69.
Zurück zum Zitat Jung K, Lein M, Stephan C, Von Hosslin K, Semjonow A, Sinha P, Loening SA, Schnorr D: Comparison of 10 serum bone turnover markers in prostate carcinoma patients with bone metastatic spread: diagnostic and prognostic implications. Int J Cancer. 2004, 111: 783-791. 10.1002/ijc.20314PubMed Jung K, Lein M, Stephan C, Von Hosslin K, Semjonow A, Sinha P, Loening SA, Schnorr D: Comparison of 10 serum bone turnover markers in prostate carcinoma patients with bone metastatic spread: diagnostic and prognostic implications. Int J Cancer. 2004, 111: 783-791. 10.1002/ijc.20314PubMed
70.
Zurück zum Zitat Keller ET, Zhang J, Cooper CR, Smith PC, McCauley LK, Pienta KJ, Taichman RS: Prostate carcinoma skeletal metastases: cross-talk between tumor and bone. Cancer Metastasis Rev. 2001, 20: 333-349. 10.1023/A:1015599831232PubMed Keller ET, Zhang J, Cooper CR, Smith PC, McCauley LK, Pienta KJ, Taichman RS: Prostate carcinoma skeletal metastases: cross-talk between tumor and bone. Cancer Metastasis Rev. 2001, 20: 333-349. 10.1023/A:1015599831232PubMed
71.
Zurück zum Zitat Liu H, Holm M, Xie XQ, Wolf-Watz M, Grundstrom T: AML1/Runx1 recruits calcineurin to regulate granulocyte macrophage colony-stimulating factor by Ets1 activation. J Biol Chem. 2004, 279: 29398-29408. 10.1074/jbc.M403173200PubMed Liu H, Holm M, Xie XQ, Wolf-Watz M, Grundstrom T: AML1/Runx1 recruits calcineurin to regulate granulocyte macrophage colony-stimulating factor by Ets1 activation. J Biol Chem. 2004, 279: 29398-29408. 10.1074/jbc.M403173200PubMed
72.
Zurück zum Zitat Park BK, Zhang H, Zeng Q, Dai J, Keller ET, Giordano T, Gu K, Shah V, Pei L, Zarbo RJ: NF-kappaB in breast cancer cells promotes osteolytic bone metastasis by inducing osteoclastogenesis via GM-CSF. Nat Med. 2007, 13: 62-69. 10.1038/nm1519PubMed Park BK, Zhang H, Zeng Q, Dai J, Keller ET, Giordano T, Gu K, Shah V, Pei L, Zarbo RJ: NF-kappaB in breast cancer cells promotes osteolytic bone metastasis by inducing osteoclastogenesis via GM-CSF. Nat Med. 2007, 13: 62-69. 10.1038/nm1519PubMed
73.
Zurück zum Zitat Grivennikov SI, Greten FR, Karin M: Immunity, inflammation, and cancer. Cell. 2010, 140: 883-899. 10.1016/j.cell.2010.01.025PubMedCentralPubMed Grivennikov SI, Greten FR, Karin M: Immunity, inflammation, and cancer. Cell. 2010, 140: 883-899. 10.1016/j.cell.2010.01.025PubMedCentralPubMed
74.
Zurück zum Zitat Dranoff G: Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer. 2004, 4: 11-22. 10.1038/nrc1252PubMed Dranoff G: Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer. 2004, 4: 11-22. 10.1038/nrc1252PubMed
75.
Zurück zum Zitat Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M: IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004, 118: 285-296. 10.1016/j.cell.2004.07.013PubMed Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M: IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004, 118: 285-296. 10.1016/j.cell.2004.07.013PubMed
76.
Zurück zum Zitat Ishii M, Egen JG, Klauschen F, Meier-Schellersheim M, Saeki Y, Vacher J, Proia RL, Germain RN: Sphingosine-1-phosphate mobilizes osteoclast precursors and regulates bone homeostasis. Nature. 2009, 458: 524-528. 10.1038/nature07713PubMedCentralPubMed Ishii M, Egen JG, Klauschen F, Meier-Schellersheim M, Saeki Y, Vacher J, Proia RL, Germain RN: Sphingosine-1-phosphate mobilizes osteoclast precursors and regulates bone homeostasis. Nature. 2009, 458: 524-528. 10.1038/nature07713PubMedCentralPubMed
77.
Zurück zum Zitat Shida D, Takabe K, Kapitonov D, Milstien S, Spiegel S: Targeting SphK1 as a new strategy against cancer. Curr Drug Targets. 2008, 9: 662-673. 10.2174/138945008785132402PubMedCentralPubMed Shida D, Takabe K, Kapitonov D, Milstien S, Spiegel S: Targeting SphK1 as a new strategy against cancer. Curr Drug Targets. 2008, 9: 662-673. 10.2174/138945008785132402PubMedCentralPubMed
78.
Zurück zum Zitat Visentin B, Vekich JA, Sibbald BJ, Cavalli AL, Moreno KM, Matteo RG, Garland WA, Lu Y, Yu S, Hall HS: Validation of an anti-sphingosine-1-phosphate antibody as a potential therapeutic in reducing growth, invasion, and angiogenesis in multiple tumor lineages. Cancer Cell. 2006, 9: 225-238. 10.1016/j.ccr.2006.02.023PubMed Visentin B, Vekich JA, Sibbald BJ, Cavalli AL, Moreno KM, Matteo RG, Garland WA, Lu Y, Yu S, Hall HS: Validation of an anti-sphingosine-1-phosphate antibody as a potential therapeutic in reducing growth, invasion, and angiogenesis in multiple tumor lineages. Cancer Cell. 2006, 9: 225-238. 10.1016/j.ccr.2006.02.023PubMed
79.
Zurück zum Zitat Pchejetski D, Golzio M, Bonhoure E, Calvet C, Doumerc N, Garcia V, Mazerolles C, Rischmann P, Teissie J, Malavaud B, Cuvillier O: Sphingosine kinase-1 as a chemotherapy sensor in prostate adenocarcinoma cell and mouse models. Cancer Res. 2005, 65: 11667-11675. 10.1158/0008-5472.CAN-05-2702PubMed Pchejetski D, Golzio M, Bonhoure E, Calvet C, Doumerc N, Garcia V, Mazerolles C, Rischmann P, Teissie J, Malavaud B, Cuvillier O: Sphingosine kinase-1 as a chemotherapy sensor in prostate adenocarcinoma cell and mouse models. Cancer Res. 2005, 65: 11667-11675. 10.1158/0008-5472.CAN-05-2702PubMed
80.
Zurück zum Zitat Sadikovic B, Thorner P, Chilton-Macneill S, Martin JW, Cervigne NK, Squire J, Zielenska M: Expression analysis of genes associated with human osteosarcoma tumors shows correlation of RUNX2 overexpression with poor response to chemotherapy. BMC Cancer. 10: 202- Sadikovic B, Thorner P, Chilton-Macneill S, Martin JW, Cervigne NK, Squire J, Zielenska M: Expression analysis of genes associated with human osteosarcoma tumors shows correlation of RUNX2 overexpression with poor response to chemotherapy. BMC Cancer. 10: 202-
81.
Zurück zum Zitat Vanhara P, Lincova E, Kozubik A, Jurdic P, Soucek K, Smarda J: Growth/differentiation factor-15 inhibits differentiation into osteoclasts--a novel factor involved in control of osteoclast differentiation. Differentiation. 2009, 78: 213-222. 10.1016/j.diff.2009.07.008PubMed Vanhara P, Lincova E, Kozubik A, Jurdic P, Soucek K, Smarda J: Growth/differentiation factor-15 inhibits differentiation into osteoclasts--a novel factor involved in control of osteoclast differentiation. Differentiation. 2009, 78: 213-222. 10.1016/j.diff.2009.07.008PubMed
82.
Zurück zum Zitat Pratap J, Lian JB, Javed A, Barnes GL, van Wijnen AJ, Stein JL, Stein GS: Regulatory roles of Runx2 in metastatic tumor and cancer cell interactions with bone. Cancer Metastasis Rev. 2006, 25: 589-600. 10.1007/s10555-006-9032-0PubMed Pratap J, Lian JB, Javed A, Barnes GL, van Wijnen AJ, Stein JL, Stein GS: Regulatory roles of Runx2 in metastatic tumor and cancer cell interactions with bone. Cancer Metastasis Rev. 2006, 25: 589-600. 10.1007/s10555-006-9032-0PubMed
83.
Zurück zum Zitat Teplyuk NM, Haupt LM, Ling L, Dombrowski C, Mun FK, Nathan SS, Lian JB, Stein JL, Stein GS, Cool SM, van Wijnen AJ: The osteogenic transcription factor Runx2 regulates components of the fibroblast growth factor/proteoglycan signaling axis in osteoblasts. J Cell Biochem. 2009, 107: 144-154. 10.1002/jcb.22108PubMedCentralPubMed Teplyuk NM, Haupt LM, Ling L, Dombrowski C, Mun FK, Nathan SS, Lian JB, Stein JL, Stein GS, Cool SM, van Wijnen AJ: The osteogenic transcription factor Runx2 regulates components of the fibroblast growth factor/proteoglycan signaling axis in osteoblasts. J Cell Biochem. 2009, 107: 144-154. 10.1002/jcb.22108PubMedCentralPubMed
84.
Zurück zum Zitat Essner JJ, Chen E, Ekker SC: Syndecan-2. Int J Biochem Cell Biol. 2006, 38: 152-156. 10.1016/j.biocel.2005.08.012PubMed Essner JJ, Chen E, Ekker SC: Syndecan-2. Int J Biochem Cell Biol. 2006, 38: 152-156. 10.1016/j.biocel.2005.08.012PubMed
85.
Zurück zum Zitat Park H, Kim Y, Lim Y, Han I, Oh ES: Syndecan-2 mediates adhesion and proliferation of colon carcinoma cells. J Biol Chem. 2002, 277: 29730-29736. 10.1074/jbc.M202435200PubMed Park H, Kim Y, Lim Y, Han I, Oh ES: Syndecan-2 mediates adhesion and proliferation of colon carcinoma cells. J Biol Chem. 2002, 277: 29730-29736. 10.1074/jbc.M202435200PubMed
86.
Zurück zum Zitat Granes F, Urena JM, Rocamora N, Vilaro S: Ezrin links syndecan-2 to the cytoskeleton. J Cell Sci. 2000, 113 (Pt 7): 1267-1276.PubMed Granes F, Urena JM, Rocamora N, Vilaro S: Ezrin links syndecan-2 to the cytoskeleton. J Cell Sci. 2000, 113 (Pt 7): 1267-1276.PubMed
87.
Zurück zum Zitat Nelson JB, Hedican SP, George DJ, Reddi AH, Piantadosi S, Eisenberger MA, Simons JW: Identification of endothelin-1 in the pathophysiology of metastatic adenocarcinoma of the prostate. Nat Med. 1995, 1: 944-949. 10.1038/nm0995-944PubMed Nelson JB, Hedican SP, George DJ, Reddi AH, Piantadosi S, Eisenberger MA, Simons JW: Identification of endothelin-1 in the pathophysiology of metastatic adenocarcinoma of the prostate. Nat Med. 1995, 1: 944-949. 10.1038/nm0995-944PubMed
88.
Zurück zum Zitat Dai J, Kitagawa Y, Zhang J, Yao Z, Mizokami A, Cheng S, Nor J, McCauley LK, Taichman RS, Keller ET: Vascular endothelial growth factor contributes to the prostate cancer-induced osteoblast differentiation mediated by bone morphogenetic protein. Cancer Res. 2004, 64: 994-999. 10.1158/0008-5472.CAN-03-1382PubMed Dai J, Kitagawa Y, Zhang J, Yao Z, Mizokami A, Cheng S, Nor J, McCauley LK, Taichman RS, Keller ET: Vascular endothelial growth factor contributes to the prostate cancer-induced osteoblast differentiation mediated by bone morphogenetic protein. Cancer Res. 2004, 64: 994-999. 10.1158/0008-5472.CAN-03-1382PubMed
89.
Zurück zum Zitat Street J, Bao M, deGuzman L, Bunting S, Peale FV, Ferrara N, Steinmetz H, Hoeffel J, Cleland JL, Daugherty A: Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc Natl Acad Sci USA. 2002, 99: 9656-9661. 10.1073/pnas.152324099PubMedCentralPubMed Street J, Bao M, deGuzman L, Bunting S, Peale FV, Ferrara N, Steinmetz H, Hoeffel J, Cleland JL, Daugherty A: Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc Natl Acad Sci USA. 2002, 99: 9656-9661. 10.1073/pnas.152324099PubMedCentralPubMed
90.
Zurück zum Zitat Cross MJ, Claesson-Welsh L: FGF and VEGF function in angiogenesis: signalling pathways, biological responses and therapeutic inhibition. Trends Pharmacol Sci. 2001, 22: 201-207. 10.1016/S0165-6147(00)01676-XPubMed Cross MJ, Claesson-Welsh L: FGF and VEGF function in angiogenesis: signalling pathways, biological responses and therapeutic inhibition. Trends Pharmacol Sci. 2001, 22: 201-207. 10.1016/S0165-6147(00)01676-XPubMed
91.
Zurück zum Zitat Ferrara N, Gerber HP, LeCouter J: The biology of VEGF and its receptors. Nat Med. 2003, 9: 669-676. 10.1038/nm0603-669PubMed Ferrara N, Gerber HP, LeCouter J: The biology of VEGF and its receptors. Nat Med. 2003, 9: 669-676. 10.1038/nm0603-669PubMed
92.
Zurück zum Zitat Vaidyanathan G, Cismowski MJ, Wang G, Vincent TS, Brown KD, Lanier SM: The Ras-related protein AGS1/RASD1 suppresses cell growth. Oncogene. 2004, 23: 5858-5863. 10.1038/sj.onc.1207774PubMed Vaidyanathan G, Cismowski MJ, Wang G, Vincent TS, Brown KD, Lanier SM: The Ras-related protein AGS1/RASD1 suppresses cell growth. Oncogene. 2004, 23: 5858-5863. 10.1038/sj.onc.1207774PubMed
93.
Zurück zum Zitat Horsch K, de Wet H, Schuurmans MM, Allie-Reid F, Cato AC, Cunningham J, Burrin JM, Hough FS, Hulley PA: Mitogen-activated protein kinase phosphatase 1/dual specificity phosphatase 1 mediates glucocorticoid inhibition of osteoblast proliferation. Mol Endocrinol. 2007, 21: 2929-2940. 10.1210/me.2007-0153PubMedCentralPubMed Horsch K, de Wet H, Schuurmans MM, Allie-Reid F, Cato AC, Cunningham J, Burrin JM, Hough FS, Hulley PA: Mitogen-activated protein kinase phosphatase 1/dual specificity phosphatase 1 mediates glucocorticoid inhibition of osteoblast proliferation. Mol Endocrinol. 2007, 21: 2929-2940. 10.1210/me.2007-0153PubMedCentralPubMed
94.
Zurück zum Zitat Sramkoski RM, Pretlow TG, Giaconia JM, Pretlow TP, Schwartz S, Sy MS, Marengo SR, Rhim JS, Zhang D, Jacobberger JW: A new human prostate carcinoma cell line, 22Rv1. In Vitro Cell Dev Biol Anim. 1999, 35: 403-409. 10.1007/s11626-999-0115-4PubMed Sramkoski RM, Pretlow TG, Giaconia JM, Pretlow TP, Schwartz S, Sy MS, Marengo SR, Rhim JS, Zhang D, Jacobberger JW: A new human prostate carcinoma cell line, 22Rv1. In Vitro Cell Dev Biol Anim. 1999, 35: 403-409. 10.1007/s11626-999-0115-4PubMed
95.
Zurück zum Zitat Hecht J, Seitz V, Urban M, Wagner F, Robinson PN, Stiege A, Dieterich C, Kornak U, Wilkening U, Brieske N: Detection of novel skeletogenesis target genes by comprehensive analysis of a Runx2(-/-) mouse model. Gene Expr Patterns. 2007, 7: 102-112. 10.1016/j.modgep.2006.05.014PubMed Hecht J, Seitz V, Urban M, Wagner F, Robinson PN, Stiege A, Dieterich C, Kornak U, Wilkening U, Brieske N: Detection of novel skeletogenesis target genes by comprehensive analysis of a Runx2(-/-) mouse model. Gene Expr Patterns. 2007, 7: 102-112. 10.1016/j.modgep.2006.05.014PubMed
96.
Zurück zum Zitat Young DW, Hassan MQ, Yang XQ, Galindo M, Javed A, Zaidi SK, Furcinitti P, Lapointe D, Montecino M, Lian JB: Mitotic retention of gene expression patterns by the cell fate-determining transcription factor Runx2. Proc Natl Acad Sci USA. 2007, 104: 3189-3194. 10.1073/pnas.0611419104PubMedCentralPubMed Young DW, Hassan MQ, Yang XQ, Galindo M, Javed A, Zaidi SK, Furcinitti P, Lapointe D, Montecino M, Lian JB: Mitotic retention of gene expression patterns by the cell fate-determining transcription factor Runx2. Proc Natl Acad Sci USA. 2007, 104: 3189-3194. 10.1073/pnas.0611419104PubMedCentralPubMed
97.
Zurück zum Zitat Wotton S, Terry A, Kilbey A, Jenkins A, Herzyk P, Cameron E, Neil JC: Gene array analysis reveals a common Runx transcriptional programme controlling cell adhesion and survival. Oncogene. 2008, 27: 5856-5866. 10.1038/onc.2008.195PubMedCentralPubMed Wotton S, Terry A, Kilbey A, Jenkins A, Herzyk P, Cameron E, Neil JC: Gene array analysis reveals a common Runx transcriptional programme controlling cell adhesion and survival. Oncogene. 2008, 27: 5856-5866. 10.1038/onc.2008.195PubMedCentralPubMed
98.
Zurück zum Zitat Shore P: A role for Runx2 in normal mammary gland and breast cancer bone metastasis. J Cell Biochem. 2005, 96: 484-489. 10.1002/jcb.20557PubMed Shore P: A role for Runx2 in normal mammary gland and breast cancer bone metastasis. J Cell Biochem. 2005, 96: 484-489. 10.1002/jcb.20557PubMed
99.
Zurück zum Zitat Khalid O, Baniwal SK, Purcell DJ, Leclerc N, Gabet Y, Stallcup MR, Coetzee GA, Frenkel B: Modulation of Runx2 activity by estrogen receptor-alpha: implications for osteoporosis and breast cancer. Endocrinology. 2008, 149: 5984-5995. 10.1210/en.2008-0680PubMedCentralPubMed Khalid O, Baniwal SK, Purcell DJ, Leclerc N, Gabet Y, Stallcup MR, Coetzee GA, Frenkel B: Modulation of Runx2 activity by estrogen receptor-alpha: implications for osteoporosis and breast cancer. Endocrinology. 2008, 149: 5984-5995. 10.1210/en.2008-0680PubMedCentralPubMed
100.
Zurück zum Zitat Javed A, Barnes GL, Pratap J, Antkowiak T, Gerstenfeld LC, van Wijnen AJ, Stein JL, Lian JB, Stein GS: Impaired intranuclear trafficking of Runx2 (AML3/CBFA1) transcription factors in breast cancer cells inhibits osteolysis in vivo. Proc Natl Acad Sci USA. 2005, 102: 1454-1459. 10.1073/pnas.0409121102PubMedCentralPubMed Javed A, Barnes GL, Pratap J, Antkowiak T, Gerstenfeld LC, van Wijnen AJ, Stein JL, Lian JB, Stein GS: Impaired intranuclear trafficking of Runx2 (AML3/CBFA1) transcription factors in breast cancer cells inhibits osteolysis in vivo. Proc Natl Acad Sci USA. 2005, 102: 1454-1459. 10.1073/pnas.0409121102PubMedCentralPubMed
101.
Zurück zum Zitat Baniwal SK, Khalid O, Sir D, Buchanan G, Coetzee GA, Frenkel B: Repression of Runx2 by androgen receptor (AR) in osteoblasts and prostate cancer cells: AR binds Runx2 and abrogates its recruitment to DNA. Mol Endocrinol. 2009, 23: 1203-1214. 10.1210/me.2008-0470PubMedCentralPubMed Baniwal SK, Khalid O, Sir D, Buchanan G, Coetzee GA, Frenkel B: Repression of Runx2 by androgen receptor (AR) in osteoblasts and prostate cancer cells: AR binds Runx2 and abrogates its recruitment to DNA. Mol Endocrinol. 2009, 23: 1203-1214. 10.1210/me.2008-0470PubMedCentralPubMed
102.
Zurück zum Zitat Morrissey C, Vessella RL: The role of tumor microenvironment in prostate cancer bone metastasis. J Cell Biochem. 2007, 101: 873-886. 10.1002/jcb.21214PubMed Morrissey C, Vessella RL: The role of tumor microenvironment in prostate cancer bone metastasis. J Cell Biochem. 2007, 101: 873-886. 10.1002/jcb.21214PubMed
103.
Zurück zum Zitat Roodman GD: Mechanisms of bone metastasis. N Engl J Med. 2004, 350: 1655-1664. 10.1056/NEJMra030831PubMed Roodman GD: Mechanisms of bone metastasis. N Engl J Med. 2004, 350: 1655-1664. 10.1056/NEJMra030831PubMed
104.
Zurück zum Zitat Mundy GR: Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer. 2002, 2: 584-593. 10.1038/nrc867PubMed Mundy GR: Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer. 2002, 2: 584-593. 10.1038/nrc867PubMed
105.
Zurück zum Zitat Logothetis CJ, Lin SH: Osteoblasts in prostate cancer metastasis to bone. Nat Rev Cancer. 2005, 5: 21-28. 10.1038/nrc1528PubMed Logothetis CJ, Lin SH: Osteoblasts in prostate cancer metastasis to bone. Nat Rev Cancer. 2005, 5: 21-28. 10.1038/nrc1528PubMed
106.
Zurück zum Zitat Phillips JE, Garcia AJ: Retroviral-mediated gene therapy for the differentiation of primary cells into a mineralizing osteoblastic phenotype. Methods Mol Biol. 2008, 433: 333-354. full_textPubMed Phillips JE, Garcia AJ: Retroviral-mediated gene therapy for the differentiation of primary cells into a mineralizing osteoblastic phenotype. Methods Mol Biol. 2008, 433: 333-354. full_textPubMed
107.
Zurück zum Zitat Kim JH, Yang CK, Heo K, Roeder RG, An W, Stallcup MR: CCAR1, a key regulator of mediator complex recruitment to nuclear receptor transcription complexes. Mol Cell. 2008, 31: 510-519. 10.1016/j.molcel.2008.08.001PubMedCentralPubMed Kim JH, Yang CK, Heo K, Roeder RG, An W, Stallcup MR: CCAR1, a key regulator of mediator complex recruitment to nuclear receptor transcription complexes. Mol Cell. 2008, 31: 510-519. 10.1016/j.molcel.2008.08.001PubMedCentralPubMed
108.
Zurück zum Zitat Benjamini Y, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B. 1995, 57: 289-300. Benjamini Y, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B. 1995, 57: 289-300.
109.
Zurück zum Zitat Toth M, Fridman R: Assessment of Gelatinases (MMP-2 and MMP-9 by Gelatin Zymography. Metastasis Research Protocols. 2008, 57: 163-174. full_text. full_text Toth M, Fridman R: Assessment of Gelatinases (MMP-2 and MMP-9 by Gelatin Zymography. Metastasis Research Protocols. 2008, 57: 163-174. full_text. full_text
110.
Zurück zum Zitat Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002, 2: 442-454. 10.1038/nrc822PubMed Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002, 2: 442-454. 10.1038/nrc822PubMed
111.
Zurück zum Zitat Dang CV, O'Donnell KA, Zeller KI, Nguyen T, Osthus RC, Li F: The c-Myc target gene network. Semin Cancer Biol. 2006, 16: 253-264. 10.1016/j.semcancer.2006.07.014PubMed Dang CV, O'Donnell KA, Zeller KI, Nguyen T, Osthus RC, Li F: The c-Myc target gene network. Semin Cancer Biol. 2006, 16: 253-264. 10.1016/j.semcancer.2006.07.014PubMed
112.
Zurück zum Zitat Guccione E, Martinato F, Finocchiaro G, Luzi L, Tizzoni L, Dall'Olio V, Zardo G, Nervi C, Bernard L, Amati B: Myc-binding-site recognition in the human genome is determined by chromatin context. Nat Cell Biol. 2006, 8: 764-770. 10.1038/ncb1434PubMed Guccione E, Martinato F, Finocchiaro G, Luzi L, Tizzoni L, Dall'Olio V, Zardo G, Nervi C, Bernard L, Amati B: Myc-binding-site recognition in the human genome is determined by chromatin context. Nat Cell Biol. 2006, 8: 764-770. 10.1038/ncb1434PubMed
113.
Zurück zum Zitat Burns JM, Summers BC, Wang Y, Melikian A, Berahovich R, Miao Z, Penfold ME, Sunshine MJ, Littman DR, Kuo CJ: A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J Exp Med. 2006, 203: 2201-2213. 10.1084/jem.20052144PubMedCentralPubMed Burns JM, Summers BC, Wang Y, Melikian A, Berahovich R, Miao Z, Penfold ME, Sunshine MJ, Littman DR, Kuo CJ: A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J Exp Med. 2006, 203: 2201-2213. 10.1084/jem.20052144PubMedCentralPubMed
114.
Zurück zum Zitat Bellou S, Hink MA, Bagli E, Panopoulou E, Bastiaens PI, Murphy C, Fotsis T: VEGF autoregulates its proliferative and migratory ERK1/2 and p38 cascades by enhancing the expression of DUSP1 and DUSP5 phosphatases in endothelial cells. Am J Physiol Cell Physiol. 2009, 297: C1477-1489. 10.1152/ajpcell.00058.2009PubMed Bellou S, Hink MA, Bagli E, Panopoulou E, Bastiaens PI, Murphy C, Fotsis T: VEGF autoregulates its proliferative and migratory ERK1/2 and p38 cascades by enhancing the expression of DUSP1 and DUSP5 phosphatases in endothelial cells. Am J Physiol Cell Physiol. 2009, 297: C1477-1489. 10.1152/ajpcell.00058.2009PubMed
115.
Zurück zum Zitat Liu F, Verin AD, Wang P, Day R, Wersto RP, Chrest FJ, English DK, Garcia JG: Differential regulation of sphingosine-1-phosphate- and VEGF-induced endothelial cell chemotaxis. Involvement of G(ialpha2)-linked Rho kinase activity. Am J Respir Cell Mol Biol. 2001, 24: 711-719.PubMed Liu F, Verin AD, Wang P, Day R, Wersto RP, Chrest FJ, English DK, Garcia JG: Differential regulation of sphingosine-1-phosphate- and VEGF-induced endothelial cell chemotaxis. Involvement of G(ialpha2)-linked Rho kinase activity. Am J Respir Cell Mol Biol. 2001, 24: 711-719.PubMed
116.
Zurück zum Zitat Liu Y, Wada R, Yamashita T, Mi Y, Deng CX, Hobson JP, Rosenfeldt HM, Nava VE, Chae SS, Lee MJ: Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J Clin Invest. 2000, 106: 951-961. 10.1172/JCI10905PubMedCentralPubMed Liu Y, Wada R, Yamashita T, Mi Y, Deng CX, Hobson JP, Rosenfeldt HM, Nava VE, Chae SS, Lee MJ: Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J Clin Invest. 2000, 106: 951-961. 10.1172/JCI10905PubMedCentralPubMed
117.
Zurück zum Zitat Donaudy F, Zheng L, Ficarella R, Ballana E, Carella M, Melchionda S, Estivill X, Bartles JR, Gasparini P: Espin gene (ESPN) mutations associated with autosomal dominant hearing loss cause defects in microvillar elongation or organisation. J Med Genet. 2006, 43: 157-161. 10.1136/jmg.2005.032086PubMedCentralPubMed Donaudy F, Zheng L, Ficarella R, Ballana E, Carella M, Melchionda S, Estivill X, Bartles JR, Gasparini P: Espin gene (ESPN) mutations associated with autosomal dominant hearing loss cause defects in microvillar elongation or organisation. J Med Genet. 2006, 43: 157-161. 10.1136/jmg.2005.032086PubMedCentralPubMed
118.
Zurück zum Zitat Bartles JR, Wierda A, Zheng L: Identification and characterization of espin, an actin-binding protein localized to the F-actin-rich junctional plaques of Sertoli cell ectoplasmic specializations. J Cell Sci. 1996, 109 (Pt 6): 1229-1239.PubMed Bartles JR, Wierda A, Zheng L: Identification and characterization of espin, an actin-binding protein localized to the F-actin-rich junctional plaques of Sertoli cell ectoplasmic specializations. J Cell Sci. 1996, 109 (Pt 6): 1229-1239.PubMed
119.
Zurück zum Zitat Gu C, Rodriguez ER, Reimert DV, Shu T, Fritzsch B, Richards LJ, Kolodkin AL, Ginty DD: Neuropilin-1 conveys semaphorin and VEGF signaling during neural and cardiovascular development. Dev Cell. 2003, 5: 45-57. 10.1016/S1534-5807(03)00169-2PubMedCentralPubMed Gu C, Rodriguez ER, Reimert DV, Shu T, Fritzsch B, Richards LJ, Kolodkin AL, Ginty DD: Neuropilin-1 conveys semaphorin and VEGF signaling during neural and cardiovascular development. Dev Cell. 2003, 5: 45-57. 10.1016/S1534-5807(03)00169-2PubMedCentralPubMed
120.
Zurück zum Zitat Singh R, Bandyopadhyay D: MUC1: a target molecule for cancer therapy. Cancer Biol Ther. 2007, 6: 481-486. 10.4161/cbt.6.4.4201PubMed Singh R, Bandyopadhyay D: MUC1: a target molecule for cancer therapy. Cancer Biol Ther. 2007, 6: 481-486. 10.4161/cbt.6.4.4201PubMed
121.
Zurück zum Zitat Tanner SL, Franzen R, Jaffe H, Quarles RH: Evidence for expression of some microtubule-associated protein 1B in neurons as a plasma membrane glycoprotein. J Neurochem. 2000, 75: 553-562. 10.1046/j.1471-4159.2000.0750553.xPubMed Tanner SL, Franzen R, Jaffe H, Quarles RH: Evidence for expression of some microtubule-associated protein 1B in neurons as a plasma membrane glycoprotein. J Neurochem. 2000, 75: 553-562. 10.1046/j.1471-4159.2000.0750553.xPubMed
122.
Zurück zum Zitat Stylli SS, Stacey TT, Verhagen AM, Xu SS, Pass I, Courtneidge SA, Lock P: Nck adaptor proteins link Tks5 to invadopodia actin regulation and ECM degradation. J Cell Sci. 2009, 122: 2727-2740. 10.1242/jcs.046680PubMedCentralPubMed Stylli SS, Stacey TT, Verhagen AM, Xu SS, Pass I, Courtneidge SA, Lock P: Nck adaptor proteins link Tks5 to invadopodia actin regulation and ECM degradation. J Cell Sci. 2009, 122: 2727-2740. 10.1242/jcs.046680PubMedCentralPubMed
123.
Zurück zum Zitat Donalies M, Cramer M, Ringwald M, Starzinski-Powitz A: Expression of M-cadherin, a member of the cadherin multigene family, correlates with differentiation of skeletal muscle cells. Proc Natl Acad Sci USA. 1991, 88: 8024-8028. 10.1073/pnas.88.18.8024PubMedCentralPubMed Donalies M, Cramer M, Ringwald M, Starzinski-Powitz A: Expression of M-cadherin, a member of the cadherin multigene family, correlates with differentiation of skeletal muscle cells. Proc Natl Acad Sci USA. 1991, 88: 8024-8028. 10.1073/pnas.88.18.8024PubMedCentralPubMed
124.
Zurück zum Zitat Hu SI, Carozza M, Klein M, Nantermet P, Luk D, Crowl RM: Human HtrA, an evolutionarily conserved serine protease identified as a differentially expressed gene product in osteoarthritic cartilage. J Biol Chem. 1998, 273: 34406-34412. 10.1074/jbc.273.51.34406PubMed Hu SI, Carozza M, Klein M, Nantermet P, Luk D, Crowl RM: Human HtrA, an evolutionarily conserved serine protease identified as a differentially expressed gene product in osteoarthritic cartilage. J Biol Chem. 1998, 273: 34406-34412. 10.1074/jbc.273.51.34406PubMed
Metadaten
Titel
Runx2 transcriptome of prostate cancer cells: insights into invasiveness and bone metastasis
verfasst von
Sanjeev K Baniwal
Omar Khalid
Yankel Gabet
Ruchir R Shah
Daniel J Purcell
Deepak Mav
Alice E Kohn-Gabet
Yunfan Shi
Gerhard A Coetzee
Baruch Frenkel
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2010
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-9-258

Weitere Artikel der Ausgabe 1/2010

Molecular Cancer 1/2010 Zur Ausgabe

Update Onkologie

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