epigenetic regulation of hgf&met receptor axis is critical ... fileopen epigenetic regulation of...

13
OPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast carcinoma Paola Bendinelli 1 , Paola Maroni 2 , Emanuela Matteucci 1 and Maria Alfonsina Desiderio* ,1 Our translational research deals with the influence of microenvironment on the phenotype and colonization of bone metastases from breast carcinoma, and on pre-metastatic niche formation. The aim of the present study was to clarify the origin of hepatocyte growth factor (HGF), ligand of Met receptor, the control of the axis HGF/Met by DNA methylation, and its importance for the nexus supportive cells-metastatic cells and for metastasis outgrowth. In bone metastasis of the 1833-xenograft model, DNA methyltransferase blockade using the chemotherapic drug 5-aza-2-deoxycytidine (decitabine) strongly reduced the expression of HGF/Met receptor axis and of E-cadherin, with decrease of metastasis wideness and osteolysis, prolonging mice survival. Thus, DNA methylation events acted as commanders of breast carcinoma cells metastatizing to bone influencing the epithelial phenotype. HGF emerged as a bone-marrow stimulus, and the exosomes seemed to furnish HGF to metastatic cells. In fact, decitabine treatment similarly affected some markers of these microvesicles and HGF, indicating that its supply to recipient cells was prevented. Notably, in bone metastasis the hypomethylation of HGF, Met and E-cadherin promoters did not appear responsible for their elevated expression, but we suggest the involvement of hypermethylated regulators and of Wwox oncosuppressor, the latter being affected by decitabine. Wwox expression increased under decitabine strongly localizing in nuclei of bone metastases. We hypothesize a role of Wwox in Met activity since in vitro Wwox overexpression downregulated the level of nuclear-Met protein fragment and Met stability, also under long exposure of 1833 cells to decitabine. HGF enhanced phosphoMet and the activity in nuclei, an effect partially prevented by decitabine. Altogether, the data indicated the importance to target the tumor microenvironment by blocking epigenetic mechanisms, which control critical events for colonization such as HGF/Met axis and Wwox, as therapy of bone metastasis. Cell Death and Disease (2017) 8, e2578; doi:10.1038/cddis.2016.403; published online 2 February 2017 The dynamic plasticity of metastases is largely dependent on epigenetic mechanisms and is responsible for opportunistic adaptations, also in the absence of DNA sequence modifications. 13 Because of the changes of microenviron- mental context, metastases need to adapt to the external cues during the various steps of the process from invasion/ dissemination until outgrowth at the secondary site. 4 Dysre- gulation of DNA methylation at CG-enriched regions (known as CpG islands or CGIs), or global genomic hypomethylation, intervenes in the plastic phenotype for successful metastatic process. Aberrant methylation has paramount consequences on tumor biology (i) for the activation of alternative transcription-start sites and the release of pluripotency loci from DNA-methylation repression, or (ii) for the blockade of transcription at methylated regions corresponding to onco- suppressors, for epigenetic silencing programs of miRNAs and of relevant adhesion molecules like E-cadherin. 1,5 Methylation of CDH1 gene with transcriptional silencing of E-cadherin, an epithelial-specific biomarker, occurs in primary breast carcinoma but not in bone metastasis. 6,7 During tumor progression, the aberrant DNA methylation might drive the phenotype switch from epithelial- mesenchymal transition (EMT) to the reverse process MET. 8 EMT promotes invasion of primary tumors like breast carcinoma, and dissemination of metastatic cells, while MET contributes to the establishment of distant skeletal metastases. 4,5 However, numerous unresolved problems regarding the molecular mechanisms underlying metastasis phenotype still remain. 9 Intermediate phenotypic changes between the two end points of epithelium and mesenchyme occur in human carcinomas, each of which is characterized by different patterns of markers and signaling pathways. 5 The growth factor network contributes to a metastable or hybrid phenotype in bone metastasis, 4 reinforcing the concept of dynamic epithelial-mesenchymal plasticity (EMP). 2 Hepato- cyte growth factor (HGF) confers to metastatic cells anoikis resistance and survival together with stem-cell characteristics through Wnt-pathway, typical of EMT phenotype, but E-cadherin-intercellular adhesion dependent on HGF would favor proliferation and drug responsiveness. 4,10,11 Bone marrow is hospitable for breast carcinoma metastases, 12 and the microenvironment stimuli may deter- mine the transcription factors and the signaling pathways activated for the EMT-MET switch in metastatic cells. 4,13,14 1 Dipartimento di Scienze Biomediche per la Salute, Molecular Pathology Laboratory, Università degli Studi di Milano, Via L. Mangiagalli 31, Milano 20133, Italy and 2 Istituto Ortopedico Galeazzi IRCCS, Via R. Galeazzi 4, Milano 20161, Italy *Corresponding author: MA Desiderio, Dipartimento di Scienze Biomediche per la Salute, Molecular Pathology Laboratory, Università degli Studi di Milano, Via L. Mangiagalli 31, Milano 20133, Italy. Tel: +39 02 50315334; Fax: +39 02 50315338; E-mail: [email protected] Received 29.7.16; revised 05.10.16; accepted 06.10.16; Edited by R Johnstone Citation: Cell Death and Disease (2017) 8, e2578; doi:10.1038/cddis.2016.403 Ofcial journal of the Cell Death Differentiation Association www.nature.com/cddis

Upload: vuongtuong

Post on 01-May-2019

217 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

OPEN

Epigenetic regulation of HGF/Met receptor axis iscritical for the outgrowth of bone metastasis frombreast carcinoma

Paola Bendinelli1, Paola Maroni2, Emanuela Matteucci1 and Maria Alfonsina Desiderio*,1

Our translational research deals with the influence of microenvironment on the phenotype and colonization of bone metastasesfrom breast carcinoma, and on pre-metastatic niche formation. The aim of the present study was to clarify the origin of hepatocytegrowth factor (HGF), ligand of Met receptor, the control of the axis HGF/Met by DNA methylation, and its importance for the nexussupportive cells-metastatic cells and for metastasis outgrowth. In bone metastasis of the 1833-xenograft model, DNAmethyltransferase blockade using the chemotherapic drug 5-aza-2′-deoxycytidine (decitabine) strongly reduced the expression ofHGF/Met receptor axis and of E-cadherin, with decrease of metastasis wideness and osteolysis, prolonging mice survival. Thus,DNA methylation events acted as commanders of breast carcinoma cells metastatizing to bone influencing the epithelialphenotype. HGF emerged as a bone-marrow stimulus, and the exosomes seemed to furnish HGF to metastatic cells. In fact,decitabine treatment similarly affected some markers of these microvesicles and HGF, indicating that its supply to recipient cellswas prevented. Notably, in bone metastasis the hypomethylation of HGF, Met and E-cadherin promoters did not appear responsiblefor their elevated expression, but we suggest the involvement of hypermethylated regulators and of Wwox oncosuppressor, thelatter being affected by decitabine. Wwox expression increased under decitabine strongly localizing in nuclei of bone metastases.We hypothesize a role of Wwox in Met activity since in vitroWwox overexpression downregulated the level of nuclear-Met proteinfragment and Met stability, also under long exposure of 1833 cells to decitabine. HGF enhanced phosphoMet and the activity innuclei, an effect partially prevented by decitabine. Altogether, the data indicated the importance to target the tumormicroenvironment by blocking epigenetic mechanisms, which control critical events for colonization such as HGF/Met axis andWwox, as therapy of bone metastasis.Cell Death and Disease (2017) 8, e2578; doi:10.1038/cddis.2016.403; published online 2 February 2017

The dynamic plasticity of metastases is largely dependent onepigenetic mechanisms and is responsible for opportunisticadaptations, also in the absence of DNA sequencemodifications.1–3 Because of the changes of microenviron-mental context, metastases need to adapt to the external cuesduring the various steps of the process from invasion/dissemination until outgrowth at the secondary site.4 Dysre-gulation of DNA methylation at CG-enriched regions (knownas CpG islands or CGIs), or global genomic hypomethylation,intervenes in the plastic phenotype for successful metastaticprocess. Aberrant methylation has paramount consequenceson tumor biology (i) for the activation of alternativetranscription-start sites and the release of pluripotency locifrom DNA-methylation repression, or (ii) for the blockade oftranscription at methylated regions corresponding to onco-suppressors, for epigenetic silencing programs of miRNAsand of relevant adhesion molecules like E-cadherin.1,5

Methylation of CDH1 gene with transcriptional silencing ofE-cadherin, an epithelial-specific biomarker, occurs in primarybreast carcinoma but not in bone metastasis.6,7

During tumor progression, the aberrant DNA methylationmight drive the phenotype switch from epithelial-

mesenchymal transition (EMT) to the reverse process MET.8

EMT promotes invasion of primary tumors like breastcarcinoma, and dissemination of metastatic cells, while METcontributes to the establishment of distant skeletalmetastases.4,5 However, numerous unresolved problemsregarding the molecular mechanisms underlying metastasisphenotype still remain.9 Intermediate phenotypic changesbetween the two end points of epithelium and mesenchymeoccur in human carcinomas, each of which is characterized bydifferent patterns of markers and signaling pathways.5 Thegrowth factor network contributes to a metastable or hybridphenotype in bone metastasis,4 reinforcing the concept ofdynamic epithelial-mesenchymal plasticity (EMP).2 Hepato-cyte growth factor (HGF) confers to metastatic cells anoikisresistance and survival together with stem-cell characteristicsthrough Wnt-pathway, typical of EMT phenotype, butE-cadherin-intercellular adhesion dependent on HGF wouldfavor proliferation and drug responsiveness.4,10,11

Bone marrow is hospitable for breast carcinomametastases,12 and the microenvironment stimuli may deter-mine the transcription factors and the signaling pathwaysactivated for the EMT-MET switch in metastatic cells.4,13,14

1Dipartimento di Scienze Biomediche per la Salute, Molecular Pathology Laboratory, Università degli Studi di Milano, Via L. Mangiagalli 31, Milano 20133, Italy and 2IstitutoOrtopedico Galeazzi IRCCS, Via R. Galeazzi 4, Milano 20161, Italy*Corresponding author: MA Desiderio, Dipartimento di Scienze Biomediche per la Salute, Molecular Pathology Laboratory, Università degli Studi di Milano, Via L.Mangiagalli 31, Milano 20133, Italy. Tel: +39 02 50315334; Fax: +39 02 50315338; E-mail: [email protected] 29.7.16; revised 05.10.16; accepted 06.10.16; Edited by R Johnstone

Citation: Cell Death and Disease (2017) 8, e2578; doi:10.1038/cddis.2016.403Official journal of the Cell Death Differentiation Association

www.nature.com/cddis

Page 2: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

HGF triggers a Twist 1 program in 1833-bonemetastatic clone,but not in MDA-MB231-invasive parental breast carcinomacells: this HGF-dependent epithelial phenotype is transient,because of the secondary activation of TGFβ1-signalingpathway related to ECMand osteolysis.15 HGF downregulatesthe nuclear co-factor Wwox leading to phosphoTwist 1 accessto nuclei, and the activation of Twist-transcription factor: whenWwox accedes into nuclei, however, unphosphorylated Twistis extruded ending its function.The so-called pre-metastatic niche can be formed by non-

tumorigenic host cells through the secretion of solublecytokines and growth factors, and by exosomes, but thetransfer of HGF from microenvironment-supportive cells tobone metastases is practically unknown.16,17 Notably, thesesmall extracellular vesicles of endocytic origin not only transferinformation from neoplastic cells to supportive cells,17 but alsothey are released by a variety of cell types into the extracellularspace or mediate direct cell-cell contacts.18,19 The primarycarcinomamay instruct the secondary site through exosomes,

as in the case of melanoma metastasis educating bone-marrow-derived cells.20 Exosomes from heparanase rich cellscontain HGF, which might influence the aggressivephenotype.21 Rab GTPases such as Rab5b of early andrecycling endosomes are commonly seen in exosomes.20,22

Some of the exosomal membrane proteins like LAMP2 are notidentified at the surface of the originating cell, consistent withcell-to-cell signaling events mediated by juxtacrine andectodomain cleavage.22

Studies are necessary to evaluate whether in breastcarcinoma cells methylation-dependent events act ascommanders of bone organotropism, and whether microen-vironment stimuli influence DNA methylation. There iscompelling evidence for the deterministic and notstochastic control of gene expression via epigeneticmechanisms,23,24 with a balance between pathways intrinsicto metastatic cells and extrinsic cues of the metastasismicroenvironment. The high expression of Met receptor insome cancers has been supposed to be consequence of DNA

Figure 1 Effect of dAza on osteolytic metastasis colonization and mice survival. (a) Representative images of three-dimensional reconstruction of μCT, and ofbioluminescence. Three xenograft mice per group were analyzed with similar results. The arrows indicate osteolysis. (b) Quantitative data for the bioluminescence signals and thepercent of inhibition due to dAza are shown. The data are the means± S.E. of five mice. ***Po0.001 versus ME. (c) Survival curve on Kaplan-Meier plots of the data from ME(n= 8) and ME+dAza (n= 12). (d and e) The means of Ki67 stained cells (percent in five fields) and of multinucleated osteoclasts (TRAP positive in 10 fields) are shown. Theassays were done in three xenograft mice. *Po0.05, ***Po0.001 versus the ME value at 15 days. ○Po0.05, ○○Po0.005 versus the corresponding ME value

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

2

Cell Death and Disease

Page 3: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

hypomethylation and histone acetylation, but the regulationmight be more complex under DNA methyltransferaseblockade.25

The aim of the present study in bone metastasis was toclarify the interplay between biological stimuli of the micro-environment and metastasis-methylation state, by examining

Figure 2 dAza affects Met and E-cadherin expression in bone metastasis of the xenograft model. We show representative images of bone metastases from mice treated ornot with dAza (xenograft mouse 1 with dAza). Five serial sections were examined for each specimen from three mice at 24 days for ME and at 24–26 days for ME plus dAzaobtaining similar results. bm, bone marrow; bo, bone; gp, growth plate; me, metastasis. Scale bar= 120 μm (reported in exemplificative panels × 20 and × 40). Negative controlsdid not show specific signals

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

3

Cell Death and Disease

Page 4: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

the effect of the blockade of DNA methyltransferases with5-aza-2′-deoxycytidine (decitabine, dAza) on the survival ofxenograft mice, and by considering the patterns of HGF/Met

receptor axis and E-cadherin. To this end, the expression ofmicroenvironmental HGFwas evaluated together with those ofexosomal Rab5b and LAMP2, to test the hypothesis that the

Figure 3 Effect of dAza on mouse and human HGF expression in bone metastasis of the xenograft model. We show representative images of bone metastases from micetreated or not with dAza (xenograft mouse 1 with dAza). Five serial sections were examined for each specimen from three mice at 24 days for ME and at 24–26 days for ME plusdAza obtaining similar results. bm, bone marrow; bo, bone; gp, growth plate; me, metastasis. Scale bar= 120 μm (reported in exemplificative panels × 20 and × 40). Negativecontrols did not show specific signals

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

4

Cell Death and Disease

Page 5: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

functionality of Met receptor in metastatic cells might dependon the ligand of mesenchymal origin. In vitro, we examined themolecular mechanisms underlying the expression and regula-tion of Met receptor by dAza and HGF, and the role played byWwox on nuclear Met function. dAza is a chemotherapic drugdirectly incorporated in the newly synthesized DNA strands,that is in phase III trial for myeloid monocyte chronicleukaemia, colangiocarcinoma and colon carcinoma,26,27 butnever tested to fight bone metastasis of women operated forprimary-breast carcinoma.The implication of HGF in the methylation-dependent

colonization of bone metastases from breast cancer mighthave a remarkable significance for therapies based on theblockade of microenvironmental stimuli.

Results

DNA-methyltransferase blockade impaired bone metas-tasis outgrowth and prolonged mice survival by affectingHGF/Met axis. For the first time we investigated themethylation-dependent control of the couple HGF ligand-Met receptor in bone metastases from breast carcinoma.Preclinical studies that test innovative therapies targeting thecross-talk microenvironmental HGF-bone metastasis arescarce,11,28 and our xenograft model exploits the 90%homology between human and mouse HGF.29

We evaluated dAza effectiveness against osteolytic-bonemetastasis (ME) in 1833-xenograft model (Figure 1a). Bymicro-computed tomography (μCT) analysis, performed onday 22 after cell injection, we observed a large reduction ofosteolytic-lesion wideness in the hind limbs under dAza(ME+dAza) in respect to ME. The ME+dAza mice survivedwithout changes of osteolysis extent until 34 days, while MEmice were dead. Bioluminescence imaging was consistentwith μCT, and in ME+dAza at 34 days the bioluminescentsignal was still lower than that of ME-22 days.Under dAza, the bioluminescence value in the hind limbs

diminished starting from early times (9 days), persistinginhibited until 22 days from 1833-cell injection (Figure 1b).See Supplementary Figure S1 for hematoxylin and eosinstaining, and for the number of mice that survived during theobservation period.Kaplan-Meier analysis showed the prolongation of

xenograft-mice survival under dAza (Figure 1c): themaximal-death incidence of ME-mice occurred between 22and 24 days, while dAza prolonged the survival until 38 days.The expression of Ki67 was examined as index of metastatic-cell proliferation:30 at 24 days, Ki67 was strongly positive inMEdecreasing of threefold under dAza; Ki67 persisted low in33–38 days dAza mice (Figure 1d). The mean value ofmultinuclear (differentiated) osteoclasts (tartrate-resistantacid phosphatase, TRAP positive) decreased in 22–24 daysdAza mice compared with ME-mice, consistent with μCT data(Figure 1e). Supplementary Figure S2 reports that dAzatreatment by reducing osteoclast differentiation (TRAP-mar-ker) preserved bone volume.To determine whether HGF/Met axis underwent DNA-

methylation control and played a crucial role in bonemetastasis outgrowth, we first studied Met pattern by

immunohistochemistry under dAza (Figure 2). In 24 daysmetastasis-bearing mice, a strongMet signal was observed allover the metastatic cells, including nuclei (see inset). UnderdAza treatment of mice 1, Met signal completely disappearedfrom bonemetastases, which seemed also strongly reduced inwideness (see Supplementary Figure S1).Since E-cadherin influences the epithelial phenotype under

HGF in bone metastasis,4,24 we studied the susceptibility ofthese adhesion molecules to dAza treatment (Figure 2).Metastatic-E-cadherin signal was downregulated by dAzaboth at plasma membrane/cytosol level and in nuclei, whichwere often positive for E-cadherin in ME.In serial sections, we examined the effect of dAza on HGFof

bone microenvironment and metastasis (Figure 3). HGF has amesenchymal origin, while Met is present in parenchymalcells,31 and bone-marrow stromal fibroblasts and megakar-yocytes produce HGF.32–34 Using anti-mouse HGFantibody, astrong HGF expression was observed in the bone marrow,even if the signal was found also in the bonemetastatic cells atcytosol and nuclear levels (Figure 3, inset).We cannot excludethat supportive cells might contribute to HGF content of bonemicroenvironment and metastasis, and dAza almost comple-tely reduced metastatic and bone-marrow signals. Asexpected, the signal obtained with anti-human HGF wasscarce in bone metastasis, and it was almost unaffected bydAza (Figure 3).Altogether, we would like to underline the paramount

importance of dAza-dependent reductions of Met signal inmetastases, and of HGF of bone marrow origin both inmicroenvironment and metastases. These findings confirmthe interaction between biological stimuli and epigeneticmechanisms like DNA methylation in bone-metastasis coloni-zation. Also, our data are in agreement with the presence of aCpG island in the human Met promoter, and of numerousmethylation sites in mouse HGF promoter (SupplementaryFigure S3). HGF internalization in bonemetastasis indicates across-talk between microenvironment and metastasis,10 butthe molecular mechanism(s) are unknown. In the followingexperiments of the present paper we will clarify some aspectsof this point.

Regulation of Met expression by dAza and HGF. Basedon the xenograft-model findings, we performed in vitrostudies with 1833-bone metastatic clone and parental MDA-MB231 cells to clarify how dAza influenced HGF/Met axis inrelation to the cell type. MDA-MB231 cells are invasive,slowly metastatizing in various organs with scarce bonetropism.10

Met 145 kDa (β subunit of the receptor) was differentlyaffected by dAza exposure in the two cell lines (Figure 4a).After 2-day dAza treatment, Met decreased in 1833 cells andincreased in MDA-MB231 cells, but Met-phosphorylation atthe catalytic site was vice versa. However, Met protein leveland phosphorylation increased concomitantly in 1833 cellsexposed to dAza-30 days, versus corresponding control c30,so that the pMet/Met ratio remained constant; in MDA-MB231cells dAza exposure for 1 day as well as for 30 days enhancedpMet/Met ratios.Then, we examined Met and its phosphorylation in

response to HGF under 2-days dAza, that reduced Met in

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

5

Cell Death and Disease

Page 6: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

1833 cells as observed in vivo (Figure 4b). PhosphoMetaugmented under the combination HGF plus dAza, comparedto the single treatments. Met protein level decreased underHGFand dAza alone in respect to untreated cells (first lane). InMDA-MB231 cells exposed to the combined treatment,dAza partly reverted the huge inhibitory effect of HGF, evenif the pMet/Met ratio was still 50% less than in untreated cells.We studied the molecular mechanisms underlying these

changes of Met protein expression in the two cell lines. In MDA-MB231 cells, the steady-state mRNA and protein levels of Met

concomitantly increased both under 2-day dAza treatment andin c30 (Figure 4c). In 1833 cells, the Met mRNA levels wereconsistent with protein levels under 1 and 2 days dAza, but indAza-30 days we did not find correspondence (Figure 4c). Toevaluate whether long-term exposure to dAza affected the Metprotein half-life in 1833 cells, we performed experiments in thepresence of cycloheximide (CHX). dAza exposure for 30 daysstrongly stabilized Met protein, in respect to dAza-1 day. Thehalf-life of Met tripled in dAza-30 days versus c30, explainingthe twofold increase in Met protein in this condition (Figure 4d).

Figure 4 Long-term dAza treatment stabilized Met protein in 1833 cells. (a and b) representative images of Western blots repeated three times are shown; vinculin was usedfor normalization. The numbers at the bottom indicate the fold variations versus the first lane, considered as 1. (c) representative images of Northern blots repeated three timesare shown; GAPDH was used for normalization. The numbers at the bottom indicate the fold variations versus the first lane, considered as 1. (d) representative images of Westernblots performed with proteins from cells treated or not with cycloheximide (CHX), in the presence or the absence of dAza; vinculin was used for normalization. The data shown inthe graphic are the means±S.E. of three independent experiments. *Po0.05, **Po0.005, ***Po0.001 versus the value at the same time of c+CHX or c30+CHX

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

6

Cell Death and Disease

Page 7: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

In 1833 cells, we deepened the knowledge of the effects ofthe long-term exposure to dAza: Met and HGF immunofluor-escence signals augmented mostly in nuclei under dAza-30days (merge images) (Figure 5a). Another key molecularcharacteristic was the HGF pattern: both the precursor and the69-kDa subunit of HGF were elevated principally under dAza-30 days (Figure 5b). Since in our experimental conditions theHGF mRNA was undetectable by Northern blot and PCR

assays, we hypothesize the provenience of HGF from theculture medium, prepared with 10% FBS, that is known tocontain growth factors. This seems in agreement withexogenous HGF origin shown in Figure 3.In conclusion, both Met and HGF expression were

highly sensible to DNA methyltransferase blockade, depend-ing on the time of exposure and the microenvironmentconditions. The reduction of HGF in the microenvironment

Figure 5 dAza effects on cellular Met distribution and HGF protein levels. (a) the dAza treated and control 1833 cells on coverslips were probed with anti-Met (green) or anti-HGF (red) antibody. The nuclei were stained with DAPI (blue). The images, taken at × 400 magnification, are representative of experiments performed in triplicate. Scalebar= 120 μm. (b) representative images of Western blots repeated three times are shown; vinculin was used for normalization. The numbers at the bottom indicate the foldvariations versus the first lane, considered as 1

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

7

Cell Death and Disease

Page 8: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

might favor the Met-dependent downregulation by dAza.Therefore, the HGF/Met axis can be suggested as target ofdAza to prevent metastasis colonization, as observed in thexenograft model.

Met activity in nuclei under HGF, and Wwox regulatoryrole. Because of the presence of Met in nuclei, that under-went HGF regulation both in xenograft-mice metastases andin 1833-metastatic cells, we evaluated some molecularmechanisms responsible for Met nuclear translocation andactivity, and for the downregulation of Met in vivo under dAza(Figures 6 and 7). In nuclei of 1833 cells Met is a 60 kDafragment;10 exogenous Wwox in tumors is known to suppressthe activity of tyrosine-kinase transcription factors by cytosolsequestering,35,36 but the role of Wwox in bone metastasishas never been investigated.As shown in Figure 6a, dAza-30 days reduced Wwox

protein level. The Met access into nuclei in a phosphorylatedform was favored by short-term and long-term dAza exposure,compared to the respective controls (Figure 6b). This was inagreement with the immunofluorescence findings for Met indAza30 versus c30 (Figure 5a). Also, Figure 6b shows thatexogenous Wwox strongly interfered with the nuclear shuttle

of Met in the presence of 2 days or 30 days dAza as well asunder basal conditions.WWOX expression vector (e.v.) tripledWwox-protein level in cytosol and nuclei, increasing also itsphosphorylation (Figure 6c). Thus, ectopic Wwox was able tointerfere with dAza effect.To clarify how WWOX e.v. interfered with Met protein

enhancement under dAza-30 days, we performed experi-ments with CHX (Figure 6d). dAza-30 days stabilized Metprotein showing a half-life of 6 h versus 2 h in c30; theblockade of new protein synthesis in cells transfected withWWOX e.v. reduced proportionally Met protein half-life.Importantly, we report the immunohistochemistry assay of

Wwox (Figure 7a). Enhancement of Wwox expression (++++)was observed under dAza throughout the metastaticcells including the nuclei; Wwox signal was practically absentin nuclei of ME, the latter presenting Wwox signal in thecytosol (++).Altogether, dAza data indicated an important role of

Wwox access into nuclei of metastatic cells, which mightreduce the level of Met also by destabilizing the protein.The scheme summarizes the changes of molecular playersunder long-term exposure to decitabine in bone metastases(Figure 7b).

Figure 6 dAza treatment and WWOX expression vector transfection on Met protein in nuclei. (a) 1833 cells exposed to dAza were used to extract total proteins that wereanalyzed byWestern blot. The experiments were repeated three times; vinculin was used for normalization. The densitometric values were used to calculate the relative variationsof Wwox protein. **Po0.005 versus the value of untreated cells (c30). (b) representative images of Western blots performed with nuclear extracts from cells exposed or not toWWOX expression vector (e.v.) and dAza; the experiments were repeated three times with similar results, and B23 was used for normalization. The numbers at the bottomindicate the fold variations versus the control, considered as 1. *Po0.05: c+WWOX e.v. versus c; c30+WWOX e.v. versus c30; dAza+WWOX e.v. versus dAza. (c) representativeimages of Western blots performed with cytosol and nuclear extracts from cells transfected withWWOX e.v.; the experiments were repeated three times with similar results, andB23 or vinculin was used for normalization. The numbers at the bottom indicate the fold variations versus the first lane, considered as 1. (d) the graphic was prepared using thedensitometric values of Western blots performed with proteins from cells treated or not with cycloheximide (CHX), in the presence or the absence of dAza30 or WWOX e.v.;vinculin was used for normalization. The data are the means± S.E. of three independent experiments. *Po0.05, **Po0.005 versus the value of c30 or dAza30. ΔPo0.05versus c30 or dAza30 under CHX at the corresponding time

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

8

Cell Death and Disease

Page 9: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

Effect of HGF on nuclear Met, and microenvironmentalorigin of HGF in bone metastasis. We further clarified theregulation of Met function in nuclei. Exogenous HGFincreased Met-GFP access to nuclei only in 1833 bone-metastatic cells (see merge) (Figure 8a); starved 1833 cellsappeared almost round-cuboid (epithelial phenotype) andMDA-MB231 triangular (mesenchymal phenotype), consis-tent with previous data.4 Noteworthy, the transactivatingactivity of Met increased in response to HGF in 1833 clone,but not in MDA-MB231 cells (Figure 8b), and dAza partlyprevented this stimulatory effect (Supplementary Figure S4).In 1833 cells, HGF increased the protein level and

phosphorylation state of endogenous-nuclear Met fragment,and Met phosphorylation persisted elevated for all theobservation period (Figure 8c). The Met fragment was presentalso in nuclei of MDA-MB231 cells, but it was less phosphory-lated than in 1833 cells, undergoing downregulation under

HGF treatment (Figure 8c). The latter finding is consistent withconfocal analysis of MDA-MB231 cells showing Met innuclei.35

The results regarding the effect of exogenousHGFonMet in1833 cells, in particular the HGF-dependent regulation ofCOOH-Met levels and activity, and the intracellular increase ofHGF under 30-days dAza, led us to clarify the microenviron-mental origin of HGF (Figure 8d). Similar to HGF, the twoexosome markers LAMP2 and Rab5b were present at highlevels in bone marrow and metastatic cells. Under dAza, notonly HGF signal practically disappeared from the twometastatic compartments, but also LAMP2 signal stronglydiminished. A relevant reduction of Rab5b expression wasshown in metastatic and supportive cells.Our data indicated that DNAmethylation state affected HGF

content of bone metastases, consistent with LAMP2 andRab5b expression, leading to suppose that exosomes might

Figure 7 Effect of dAza on Wwox expression in bone metastasis of the xenograft model (ME). (a) we show representative images of bone metastases from mice treated ornot with dAza (xenograft mouse 3 with dAza). Five serial sections were examined for each specimen from three mice at 24 days for ME and at 24–26 days for ME plus dAzaobtaining similar results. bm, bone marrow; gp, growth plate; me, metastasis; bo, bone. Scale bar= 120 μm (reported in exemplificative panels × 20 and × 60). Negative controlsdid not show specific signals. (b) we show a schematic representation of the changes of the principal players occurring under dAza in bone metastases

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

9

Cell Death and Disease

Page 10: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

originate from supportive cells permitting HGF access torecipient cells in the metastatic bulk with redistribution invarious compartments.

DiscussionHost cells through the release of biological stimuli like HGFseem to support metastasis colonization, as testified by

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

10

Cell Death and Disease

Page 11: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

DNA-methyltransferase blockade with decitabine (dAza) whichhampered the HGF (mouse)/Met (human) axis. Notably, themicroenvironmental HGFboundMet at plasma-membrane leveland in nuclei of metastatic cells. Consequently, under dAza themice survival was prolonged due to the reduction of metastaticcell proliferation, with decreases of metastasis wideness andosteolysis. Our data were consistent with HGF production bybone marrow cells,32–34 and with an extensive autoactivation ofthe HGF precursor stored in the stroma via proteolysis.31 AnHGF-dependent downregulation ofmiR-27b and upregulation ofthe target ST14/matriptase is proposed.37 Since HGF sub-stitutes for M-CSF to support osteoclastogenesis,38,39 decita-bine treatment might intervene halting this mechanism andreducing osteolysis in bone metastasis. HGF is known to triggerTGFβ1-signaling pathway also involved in osteoclastactivation4,15 and to activate Twist,4 which facilitates establish-ment of osteolytic metastases through miR-10b.40

Interestingly, DNA-methyltransferase blockade reducedHGF access to metastatic cells, and various molecular eventsmight be responsible. HGF was likely to be supplemented tometastatic cells by exosomes, which might originate fromsupportive cells like megakaryocytes18 presenting high HGFlevels.34 In fact, in bone-metastatic tissue under decitabine theexosome marker LAMP2 and the mouse HGF expressionconcomitantly decreased. The exosomes may influence themethylation state of metastasis and microenvironment indifferent manners:41 many of the proteins and miRNAscontained in exosomes are regulators of DNA methyltrans-ferases or of cytidine deaminase, an enzyme involved in DNAinitiative demethylation. As a consequence, Met and E-cad-herin might be reduced in bone metastasis of the xenograftmodel by direct or indirect mechanisms. Moreover, LAMP2promoter has numerous putative sites for methylation,42

participating in LAMP2-protein expression. In bone metastatictissue, LAMP2 might be responsible for the formation ofexosomes, implicated in juxtacrine communication withinmetastasis area,22 but also for autolysosome activity beingLAMP2 present in the lysosome membranes.43 A LAMP2-mediated degradation process cannot be excluded under ourexperimental conditions, since autophagy is prometastatic byfuelling nutrients and supplying energy.44,45 Rab5b intervenesin the early endosomal phase, that precedes exosomeformation, and it was largely affected by DNA-methyltransferase blockade.For the first time, we demonstrated the interaction between

HGFand epigenetic mechanisms controlling gene expression,important for metastasis phenotype. It might be easier toprevent bone-metastasis colonization by modifying the

HGF-dependent MET (epithelial) phenotype - testified by thedecrease of E-cadherin expression under decitabine - ratherthan to change the genotype. The plasticity of bonemetastasislargely depends on phenotype adjustment,1–3 and Twistexpression also diminishes at the invasive front of bonemetastases in the 1833-xenograft model under dAza.46

Commonalities between in vivo and in vitro conditionsregard the indirect effect of dAza on HGF: the lack of HGFmRNA in 1833 cells excluded dAza effect on the promoter,while the hampering of HGF supply can be hypothesized. Inagreement, exogenous HGF seemed to play a pivotal role byreverting the inhibitory effect of dAza on Met protein level andnuclear COOH-Met activity.Wwox accumulation seemed critical for the nuclear COOH-

Met decrease under long-term exposure to dAza in thexenograft mice, probably because it prevented the access tonuclei as shown in vitro. Similarly, Wwox prevents the nucleartranslocation of Twist.4 In addition, we observed that Wwoxreduced the half-life of cellular Met, further explaining why inthe xenograft model Met signal diminished under dAza. Thus,the discrepancy between in vivo and in vitro data was onlyapparent, because in 1833 cells dAza actually decreasedWwox preventing Met destabilization, while in the xenograftmice dAza accumulated Wwox.The regulation of Met expression is complex, and the effect

of decitabine cannot be explained by hypomethylation of Metpromoter in bone metastasis. In various types of tumors,regulatory sequences intervene in Met expression such as theantisense L1-Met, that is hypermethylated and is inverselycorrelated with the transcript and the protein level of Met andwith metastasis; also the promoter of long non-coding RNAgene 3 (IntRNA Meg3) is hypermethylated favoring Metexpression, while under decitabine Met downregulationoccurs.25

Only in 1833 cells but not in parental MDA-MB231 cells,HGF activated the cytosol-nuclear shuttle of COOH-Metfragments and the Met-receptor transactivation. Nuclear Metmight transactivate genes for bonemetastatization under HGFstimulus, and the decrease of HGF due to decitabine waslikely to influence the phenotypic changes. As shown in ourprevious preclinical study, an effective anti-metastatic treat-ment was the combination NK4 plus dasatinib, inhibitors ofHGF and Src activity.11 The interactive crosstalk between Metand EGFR/HER2/VEGFR is responsible for Met resistance totyrosine-kinase receptor targeted therapies,47 and the lack ofstandardized biomarkers predictive for patient responsereduces the benefits.48 We add insight that new effectiveapproaches would target the microenvironment of bone

Figure 8 HGF treatment on Met protein and activity in nuclei, and effect of dAza on the expression of mouse HGF, LAMP2 and Rab5b in bone metastasis of the xenograftmodel. (a) cells were transfected with Met-GFP on coverslips, and exposed to HGF under starvation. The nuclei were stained with DAPI (blue). The images, taken at × 400magnification, are representative of experiments performed in triplicate. (b) the cells were co-transfected with the construct containing Met fused with Gal4 DNA-binding domain,and the reporter plasmid encoding Firefly luciferase controlled by Gal4-responsive promoter (Gal4Luc). The data are the means±S.E. of three independent experimentsperformed in triplicate. *Po0.05, **Po0.005 versus Gal4Luc transfected cells. (c) representative images of Western blots performed with nuclear extracts from cells exposed toHGF; the experiments were repeated three times, and B23 was used for normalization. The numbers at the bottom indicate the fold variations versus the first lane, consideredas 1. (d) we show representative images of bone metastases from mice treated or not with dAza (xenograft mouse 1 with dAza). Five serial sections were examined for eachspecimen from three mice at 24 days for ME and at 24–26 days for ME plus dAza with similar results. bm, bone marrow; me, metastasis; bo, bone. Scale bar= 120 μm (reportedin exemplificative panels × 20 and × 40). Negative controls did not show specific signals

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

11

Cell Death and Disease

Page 12: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

metastases, formed by bone marrow cells and a combinationof biological components.49

HGF/Met axis and Wwox are critical molecular playersregulated by DNA methyltransferases, which are important forepithelial phenotype and colonization of bone metastasis frombreast carcinoma. The therapeutic approaches targeting thenexus microenvironment-DNA methylation of metastatic cells,with consequent defects in the biological stimuli and adhesivemolecules, are promising to prevent colonization of osteolyticbone metastases affecting also the pre-metastatic nicheformation.46

Materials and MethodsCells. 1833-bone metastatic clone, the parental MDA-MB231 breast carcinomacells, and the 1833-retrovirally transfected with HSV1-tk/GFP/firefly luciferase (1833/TGL) were kindly furnished by Dr. J. Massagué (Memorial Sloan-Kettering CancerCenter, New York, USA). The cells, routinely maintained in DMEM containing 10%FBS (Sigma-Aldrich, Saint Louis, MO, USA), were used after 2 or 3 passages inculture.11

Xenograft model preparation. Animal studies were carried out accordingto the Institutional Guide for Care and Use of Laboratory Animals, and withInternational Laws. The 1833/TGL cells were injected in the heart of anaesthetizednu/nu mice: one animal group (ME, n= 8) received only the 1833/TGL cells; asecond group (ME+dAza, n= 12) received 1833/TGL cells concomitant with 5 μg/gof dAza i.p., that was given once a week until the suppression.11,46 The animalswere killed to prevent the suffering.

Immunohistochemistry. Bone samples were fixed, decalcified and serialsections were prepared.11 Immunostaining was performed with anti-Met (C-12,1 :100) (Santa Cruz Biotechnology, Santa Cruz, CA, USA), anti-mouse HGF (10 μg/ml) (R&D Systems, Minneapolis, MN, USA), anti-human αHGF (H487, 3 μg/ml)(Immuno-Biological Laboratories, Aramachi, Takasaki-Shi, Gunma, Japan), anti-E-cadherin (1 : 100) (BD Transduction Laboratories, Lexington, KY, USA), anti-LAMP2(ABL-93, 1 : 50) (Santa Cruz Biotechnology), anti-Rab5b (A-20, 1 : 50) (Santa CruzBiotechnology) or anti-Wwox (1 : 200) (Imgenex, San Diego, CA, USA) antibody.Anti-Ki67 antibody (Clone SP6, 1 : 200) was from Spring Bioscience (Pleasanton,CA, USA); after immunostaining, the mean proportion of positive cells wascalculated from any five fields by examining metastasis sections under microscopeat high magnification (×400). The Ki67-positive metastatic cells exhibited brownpunctuate granules in the nucleus, and occasionally a weak signal occurred in thecytoplasm. The in situ detection of osteoclasts was performed on TRAP-stainedparaffin-embedded medial sections of tibial metaphysics with the use of acommercial kit (Sigma-Aldrich), and the mean of multinucleated osteoclasts in 10fields was calculated after observation under microscope at high magnification.

Western blot analyses. Total (100 μg) or nuclear (50 μg) protein extractswere prepared from cells treated as follows. (i) The cells were exposed to 1 μMdAza for 1 or 2 days (24 h apart), in the presence or the absence of HGF 200 ng/ml.4,50 For HGF treatment, the cells were starved overnight. For 30 days dAzatreatment, 0.1 μM dAza was added to the cells every three days50; correspondingcontrol cells (c30) were carried out. (ii) Some cells were transfected with 400 ng/mlof WWOX e.v. (S. Semba, Kobe University, Japan)4 during the last 1 day of the dAzatreatments. (iii) Some cells exposed to 1 day or 30 days dAza, and the respectivecontrols, were treated with CHX (100 μg/ml) for various times,6 and the Westernblots were performed with total or nuclear protein extracts. Primary antibodydilutions were: anti-Met (C-12, 1 : 200) (Santa Cruz Biotechnology), anti-phosphoMet 1234/35 (catalytic site) (1 : 2000) (Transduction Laboratories), anti-αHGF (C20, 1.5 μg/ml) (Santa Cruz Biotechnology), anti-Wwox (N19, 1 : 200)(Santa Cruz Biotechnology) and anti-phosphoWwox (1 : 500) (Abcam, Cambridge,UK). Densitometric analysis was performed after reaction with ECL pluschemiluminescence kit from Thermo-Fisher Scientific (Waltham, MA, USA).

mRNA evaluation. Total RNA (30 μg) was analyzed by Northern blot, and thefilters were hybridized with the labeled human cDNAs for Met (0.84 kb) and HGF(2.2 kb) kindly given by R. Zarnegar (University of Pittsburgh, PA). Reversetranscription was performed using SuperScript VILO cDNA Synthesis Kit

(Invitrogen, Monza, Italy). Gene expression was measured using TaqMan Geneexpression assay (ThermoFisher Scientific; HGF, assay ID Hs00300159_m1).46

GAPDH was used as internal control to normalize the mRNA levels.

Immunofluorescence assay. The cells (4 × 104) on coverslips were probedwith anti-Met or anti-αHGF antibody (1 : 50). The images were collected underEclipse 80i, Nikon Fluorescence microscope at × 400 magnification.4

Transfection with Met fluorescent constructs. The cells on coverslipswere transfected with 200 ng/well of Met-expression vector, labeled with GFP,35

subcloned from pMOG-Met (full length Met cDNA) kindly given by L. Schmidt(National Cancer Institute, Frederick, MD, USA). Starved cells were treated with100 ng/ml HGF for 24 h. The images were collected under Eclipse 80i, NikonFluorescence microscope at × 400 magnification. To measure Met activity, the cellsin 24-multiwell plates were co-transfected with 200 ng of the construct containingMet fused with Gal4BD, and with 200 ng of G5-38HIV-Luc (Gal4Luc), encodingFirefly luciferase controlled by a Gal4-responsive promoter; these cells wereexposed or not to HGF for 1 day.35 The transfection was performed with a DNA:Fugene 6 mixture (3 : 1), containing the internal control phRG-TK vector(Renilla-luciferase gene), and the Firefly/Renilla luciferase activity ratios werecalculated by the software.

Statistical analysis. The statistical analysis of the densitometric andluciferase values was performed by analysis of variance. The survival data ofxenograft mice were analyzed by Kaplan-Meier method and the Log-rank (Mantel-Cox) test. Po0.05 was considered significant.

Conflict of InterestThe authors declare no conflict of interest.

Acknowledgements. Grant support was provided by Università degli Studi diMilano, Piano di Sviluppo dell’Ateneo-UNIMI 2015-linea 2 to M.A.D. and Ministerodella Salute, Ricerca Corrente (L4069, L4071, L4077) to P.M.

Author contributionsConception and design: MAD; development of methodology: MAD, PB, PM and EM;acquisition of data: PM, EM and P.B.; analysis and interpretation of data: MAD, PBand PM; writing of the manuscript: MAD; administrative, technical, or materialsupport: MAD and PM; study supervision: MAD

1. Vizoso M, Esteller M. DNA methylation plasticity contributes to the natural history ofmetastasis. Cell Cycle 2015; 14: 2863–2864.

2. Ferrao PT, Behren A, Anderson RL, Thompson EW. Editorial: cellular and phenotypicplasticity in cancer. Front Oncol 2015; 5: 171.

3. Ye X, Weinberg RA. Epithelial-mesenchymal plasticity: a central regulator of cancerprogression. Trends Cell Biol 2015; 25: 675–686.

4. Bendinelli P, Maroni P, Matteucci E, Desiderio MA. HGF and TGFβ1 differently influencedWwox regulatory function on Twist program for mesenchymal-epithelial transition in bonemetastatic versus parental breast carcinoma cells. Mol Cancer 2015; 14: 112.

5. Nickel A, Stadler SC. Role of epigenetic mechanisms in epithelial-to-mesenchymal transitionof breast cancer cells. Transl Res 2015; 165: 126–142.

6. Maroni P, Matteucci E, Drago L, Banfi G, Bendinelli P, Desiderio MA. Hypoxia inducedE-cadherin involving regulators of Hippo pathway due to HIF-1α stabilization/nucleartranslocation in bone metastasis from breast carcinoma. Exp Cell Res 2015; 330: 287–299.

7. Gunasinghe NP, Wells A, Thompson EW, Hugo HJ. Mesenchymal-epithelial transition(MET) as a mechanism for metastatic colonisation in breast cancer. Cancer Metastasis Rev2012; 31: 469–478.

8. Davalos V, Moutinho C, Villanueva A, Boque R, Silva P, Carneiro F et al. Dynamic epigeneticregulation of the microRNA-200 family mediates epithelial and mesenchymal transitions inhuman tumorigenesis. Oncogene 2012; 31: 2062–2074.

9. Bill R, Christofori G. The relevance of EMT in breast cancer metastasis: correlation orcausality? FEBS Lett 2015; 589: 1577–1587.

10. Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interactionbetween human-breast cancer metastasis and bone microenvironment through activatedhepatocyte growth factor/Met and beta-catenin/Wnt pathways. Eur J Cancer 2010; 46:1679–1691.

11. Maroni P, Bendinelli P, Matteucci E, Locatelli A, Nakamura T, Scita G et al. Osteolytic bonemetastasis is hampered by impinging on the interplay among autophagy, anoikis andossification. Cell Death Dis 2014; 5: e1005.

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

12

Cell Death and Disease

Page 13: Epigenetic regulation of HGF&Met receptor axis is critical ... fileOPEN Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast

12. Psaila B, Lyden D, Roberts I. Megakaryocytes, malignancy and bone marrowvascular niches. J Thromb Haemost 2012; 10: 177–188.

13. Tam WL, Weinberg RA. The epigenetics of epithelial-mesenchymal plasticity in cancer.Nat Med 2013; 19: 1438–1449.

14. Mani SA. Mesenchyme Forkhead 1 (FOXC2) plays a key role in metastasis and is associatedwith aggressive basal-like breast cancers. Proc Natl Acad Sci USA 2007; 104: 10069–10074.

15. Kakonen SM, Selander KS, Chirgwin JM, Yin JJ, Burns S, Rankin WA et al. Transforminggrowth factor-beta stimulates parathyroid hormone-related protein and osteolytic metastasesvia Smad and mitogen-activated protein kinase signaling pathways. J Biol Chem 2002; 277:24571–24578.

16. Massagué J, Obenauf AC. Metastatic colonization by circulating tumor cells. Nature 2016;529: 298–306.

17. Vella LJ. The emerging role of exosomes in epithelial–mesenchymal-transition in cancer.Front Oncol 2014; 4: 361.

18. Aoki J, Ohashi K, Mitsuhashi M, Murakami T, Oakes M, Kobayashi T et al.Postransplantation bone marrow assessment by quantifying hematopoietic cell-derivedmRNAs in plasma exosomes/microvesicles. Clin Chem 2014; 60: 675–682.

19. Ge M, Ke R, Cai T, Yang J, Mu X. Identification and proteomics analysis of osteoblast-derivedexosomes. Biochem Biophys Res Commun 2015; 467: 27–32.

20. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G et al.Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastaticphenotype through MET. Nat Med 2012; 18: 883–891.

21. Thompson CA, Purushothaman A, Ramani VC, Vlodavsky I, Sanderson RD. Heparanaseregulates secretion, composition, and function of tumor cell-derived exosomes. J Biol Chem2013; 288: 10093–10099.

22. Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important inintercellular communication. J Proteomics 2010; 73: 1907–1920.

23. Dumont N, Wilson MB, Crawford YG, Reynolds PA, Sigaroudinia M, Tlsty TD.Sustained induction of epithelial to mesenchymal transition activates DNA methylation ofgenes silenced in basal-like breast cancer. Proc Natl Acad Sci USA 2008; 105:14867–14872.

24. Matteucci E, Maroni P, Luzzati A, Perrucchini G, Bendinelli P, Desiderio MA. Bone metastaticprocess of breast cancer involves methylation state affecting E-cadherin expression throughTAZ and WWOX nuclear effectors. Eur J Cancer 2013; 49: 231–244.

25. Zhang J, Babic A. Regulation of the Met oncogene: molecular mechanisms. Carcinogenesis2016; 37: 345–355.

26. Nagaraju GP, El-Rayes BF. SPARC and DNA methylation: possible diagnostic andtherapeutic implications in gastrointestinal cancers. Cancer Lett 2013; 328: 10–17.

27. Navada SC, Steinmann J, Lübbert M, Silverman LR. Clinical development of demethylatingagents in hematology. J Clin Invest 2014; 124: 40–46.

28. Liu W, Kovacevic Z, Peng Z, Jin R, Wang P, Yue F et al. The molecular effect of metastasissuppressors on Src signaling and tumorigenesis: new therapeutic targets. Oncotarget 2015;6: 35522–35541.

29. Liu Y, Michalopoulos GK, Zarnegar R. Molecular cloning and characterization of cDNAencoding mouse hepatocyte growth factor. Biochim Biophys Acta 1993; 1216: 299–303.

30. Li H, Han X, Liu Y, Liu G, Dong G. Ki67 as a predictor of poor prognosis in patients with triple-negative breast cancer. Oncol Lett 2015; 9: 149–152.

31. Michalopoulos GK. Hepatocyte growth factor (HGF) and its receptor (Met) in liverregeneration, neoplasia and disease. In: Jirtle RL (ed). Liver Regeneration andCarcinogenesis. Molecular and Cellular Mechanisms. Academic Press: San Diego, CA,1995, pp 27-49.

32. Abed E, Bouvard B, Martineau X, Jouzeau JY, Reboul P, Lajeunesse D. Elevated hepatocytegrowth factor levels in osteoarthritis osteoblasts contribute to their altered response to bonemorphogenetic protein-2 and reduced mineralization capacity. Bone 2015; 75: 111–119.

33. Chowdhury R, Webber JP, Gurney M, Mason MD, Tabi Z, Clayton A. Cancer exosomestrigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasivemyofibroblasts. Oncotarget 2015; 6: 715–731.

34. Bendinelli P, Maroni P, Matteucci E, Desiderio MA. Hypoxia and microenvironment crosstalkin bone metastasis. Int J Mol Sci 2016; 17: pii E706.

35. Matteucci E, Bendinelli P, Desiderio MA. Nuclear localization of active HGF receptorMet in aggressive MDA-MB231 breast carcinoma cells. Carcinogenesis 2009; 30:937–945.

36. Aqeilan RI, Donati V, Palamarchuk A, Trapasso F, Kaou M, Pekarsky Y et al. WW-domaincontaining proteins, WWOX and YAP, compete for interaction with ErbB4 and modulate itstranscriptional function. Cancer Res 2005; 65: 6764–6772.

37. Susuki D, Kimura S, Naganuma S, Tsuchiyama K, Tanaka T, Kitamura N et al. Regulation ofmicroRNA expression by hepatocyte growth factor in human head and neck squamous cellcarcinoma. Cancer Sci 2011; 102: 2164–2171.

38. Adamopoulos IE, Xia Z, Lau YS, Athanasou NA. Hepatocyte growth factor can substitute forM-CSF to support osteoclastogenesis. Biochem Biophys Res Commun 2006; 350: 478–483.

39. Taylor RM, Kashima TG, Knowles HJ, Athanasou NA. VEGF, FLT3 ligand, PIGF and HGFcan substitute for M-CSF to induce human osteoclast formation: implications for giant celltumour pathobiology. Lab Invest 2012; 92: 1398–1406.

40. Croset M, Goehrig D, Frackowiak A, Bonnelye E, Ansieau S, Puisieux A et al. TWIST1expression in breast cancer cells facilitates bone metastasis formation. J Bone Miner Res2014; 29: 1886–1899.

41. Qian Z, Shen Q, Yang X, Qiu Y, Zhang W. The role of extracellular vesicles: an epigeneticview of the cancer microenvironment. Biomed Res Int 2015; 649161–649168.

42. Manoni M, Tribioli C, Lazzari B, DeBellis G, Patrosso C, Pergolizzi R et al. The nucleotidesequence of a CpG island demonstrates the presence of the first exon of the gene encodingthe human lysosomal membrane protein lamp2 and assigns the gene to Xq24. Genomics1991; 9: 551–554.

43. Wei SH, Li W, Liu Y, Gao DK, Pan J, Gu CW et al. Disturbance of autophagy-lysosomesignaling molecule expression in human gastric adenocarcinoma. Oncol Lett 2014; 7:635–640.

44. Maroni P, Bendinelli P, Resnati M, Matteucci E, Milan E, Desiderio MA. The autophagicprocess occurs in human bone metastasis and implicates molecular mechanisms differentlyaffected by Rab5a in the early and late stages. Int J Mol Sci 2016; 17: pii E443.

45. Panda PK, Mukhopadhyay S, Das DN, Sinha N, Naik PP, Bhutia SK. Mechanism ofautophagic regulation in carcinogenesis and cancer therapeutics. Semin Cell Dev Biol 2015;39: 43–55.

46. Matteucci E, Maroni P, Disanza A, Bendinelli P, Desiderio MA. Coordinate regulation ofmicroenvironmental stimuli and role of methylation in bone metastasis from breastcarcinoma. Biochim Biophys Acta 2016; 1863: 64–76.

47. Maroun CR, Rowlands T. The Met receptor tyrosine kinase: a key player in oncogenesis anddrug resistance. Pharmacol Ther 2014; 142: 316–338.

48. Zhang Y, Du Z, Zhang M. Biomarker development in MET-targeted therapy. Oncotarget2016; 7: 37370–37389.

49. Kaplan RN, Psaila B, Lyden D. Bone marrow cells in the “pre-metastatic niche”: within boneand beyond. Cancer Metastasis Rev 2006; 25: 521–529.

50. Kastl L, Brown I, Schofield AC. Altered DNA methylation is associated with docetaxelresistance in human breast cancer cells. Int J Oncol 2010; 36: 1235–1241.

Cell Death and Disease is an open-access journalpublished by Nature Publishing Group. This work is

licensed under a Creative Commons Attribution 4.0 InternationalLicense. The images or other third party material in this article areincluded in the article’s Creative Commons license, unless indicatedotherwise in the credit line; if the material is not included under theCreative Commons license, users will need to obtain permission fromthe license holder to reproduce the material. To view a copy of thislicense, visit http://creativecommons.org/licenses/by/4.0/

r The Author(s) 2017

Supplementary Information accompanies this paper on Cell Death and Disease website (http://www.nature.com/cddis)

Metastatic HGF/Met axis control by DNA methylationP Bendinelli et al

13

Cell Death and Disease