Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
2.
Mol Cancer ; 19(1): 126, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32799870

ABSTRACT

This article has been retracted. Please see the retraction notice for more detail: https://doi.org/10.1186/s12943-015-0389-y.

4.
Int J Mol Sci ; 20(10)2019 May 22.
Article in English | MEDLINE | ID: mdl-31121879

ABSTRACT

Hepatocyte growth factor (HGF) and transforming growth factor ß1 (TGFß1) are biological stimuli of the micro-environment which affect bone metastasis phenotype through transcription factors, but their influence on the growth is scarcely known. In a xenograft model prepared with 1833 bone metastatic cells, derived from breast carcinoma cells, we evaluated mice survival and Twist and Snail expression and localization after competitive inhibition of HGF with NK4, or after blockade of TGFß1-type I receptor (RI) with SB431542: in the latter condition HGF was also measured. To explain the in vivo data, in 1833 cells treated with SB431542 plus TGFß1 we measured HGF formation and the transduction pathway involved. Altogether, HGF seemed relevant for bone-metastatic growth, being hampered by NK4 treatment, which decreased Twist more than Snail in the metastasis bulk. TGFß1-RI blockade enhanced HGF in metastasis and adjacent bone marrow, while reducing prevalently Snail expression at the front and bulk of bone metastasis. The HGF accumulation in 1833 cells depended on an auxiliary signaling pathway, triggered by TGFß1 under SB431542, which interfered in the transcription of HGF activator inhibitor type 1 (HAI-1) downstream of TGFß-activated kinase 1 (TAK1): HGF stimulated Twist transactivation. In conclusion, the impairment of initial outgrowth with NK4 seemed therapeutically promising more than SB431542 chemotherapy; a functional correlation between Twist and Snail in bone metastasis seemed to be influenced by the biological stimuli of the micro-environment, and the targeting of these phenotype biomarkers might inhibit metastasis plasticity and colonization, even if it would be necessary to consider the changes of HGF levels in bone metastases undergoing TGFß1-RI blockade.


Subject(s)
Benzamides/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Dioxoles/therapeutic use , Hepatocyte Growth Factor/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Animals , Bone Neoplasms/pathology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Mice
5.
Cell Death Dis ; 9(5): 472, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29700305

ABSTRACT

Bone is the most common site for breast cancer spread. In the pro-metastatic cell line 1833, derived from MDA-MB-231 breast adenocarcinoma cells, both hypoxia and hepatocyte growth factor (HGF) influence the effect of miR-125b on ETS proto-oncogene 1 transcription factor (ETS1). The effect of hypoxia inducible factor 1 alpha subunit (HIF1A), known to promote metastatic spread by upregulating prostaglandin endoperoxide synthase 2 (PTGS2), may be dampened by miR-125b targeting PTGS2. Here, we investigated whether miR-125b plays a role in breast cancer metastasis by measuring its activity in response to the chemotherapeutic agent NS-398 in a xenograft model. NS-398 is typically used in the clinic to target PTGS2. We also aimed to describe the molecular mechanisms in vitro, since the enhancement of epithelial properties may favor the efficacy of therapies. We report that in the xenograft model, miR-125b reduced metastasis to the bone. We also report suppression of PTGS2 enhanced survival by decreasing HIF1A in cells within the bone marrow. In 1833 cells transfected with a miR-125b mimic we observed several phenotypic changes including enhancement of the epithelial marker E-cadherin, a reduction of mesenchymal-associated genes and a reduction of WNT-associated stem cell signaling. Our findings suggest that in vivo, key players of the bone microenvironment promoting breast cancer spread are regulated by miR-125b. In future, biological molecules imitating miR-125b may enhance the sensitivity of chemotherapeutic agents used to counteract bone metastases.


Subject(s)
Adenocarcinoma/drug therapy , Bone Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Cyclooxygenase 2/metabolism , MicroRNAs/metabolism , Nitrobenzenes/pharmacology , Proto-Oncogene Protein c-ets-1/metabolism , RNA, Neoplasm/metabolism , Sulfonamides/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclooxygenase 2/genetics , Female , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasm Metastasis , Proto-Oncogene Mas , Proto-Oncogene Protein c-ets-1/genetics , RNA, Neoplasm/genetics , Xenograft Model Antitumor Assays
6.
Int J Mol Sci ; 19(1)2018 Jan 16.
Article in English | MEDLINE | ID: mdl-29337876

ABSTRACT

We examined the influence of microenvironment stimuli on molecular events relevant to the biological functions of 1833-bone metastatic clone and the parental MDA-MB231 cells. (i) In both the cell lines, hepatocyte growth factor (HGF) and the osteoblasts' biological products down regulated nuclear Ets-1-protein level in concomitance with endogenous miR-125b accumulation. In contrast, under hypoxia nuclear Ets-1 was unchanged, notwithstanding the miR-125b increase. (ii) Also, the 1833-cell invasiveness and the expression of Endothelin-1, the target gene of Ets-1/HIF-1, showed opposite patterns under HGF and hypoxia. We clarified the molecular mechanism(s) reproducing the high miR-125b levels with the mimic in 1833 cells. Under hypoxia, the miR-125b mimic maintained a basal level and functional Ets-1 protein, as testified by the elevated cell invasiveness. However, under HGF ectopic miR-125b downregulated Ets-1 protein and cell motility, likely involving an Ets-1-dominant negative form sensible to serum conditions; Ets-1-activity inhibition by HGF implicated HIF-1α accumulation, which drugged Ets-1 in the complex bound to the Endothelin-1 promoter. Altogether, 1833-cell exposure to HGF would decrease Endothelin-1 transactivation and protein expression, with the possible impairment of Endothelin-1-dependent induction of E-cadherin, and the reversion towards an invasive phenotype: this was favoured by Ets-1 overexpression, which inhibited HIF-1α expression and HIF-1 activity. (iii) In MDA-MB231 cells, HGF strongly and rapidly decreased Ets-1, hampering invasiveness and reducing Ets-1-binding to Endothelin-1 promoter; HIF-1α did not form a complex with Ets-1 and Endothelin-1-luciferase activity was unchanged. Overall, depending on the microenvironment conditions and endogenous miR-125b levels, bone-metastatic cells might switch from Ets-1-dependent motility towards colonization/growth, regulated by the balance between Ets-1 and HIF-1.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Hepatocyte Growth Factor/pharmacology , MicroRNAs/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Tumor Microenvironment , Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , DNA/metabolism , Endothelin-1/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/genetics , Models, Biological , Neoplasm Invasiveness , Transcriptional Activation/drug effects , Tumor Microenvironment/drug effects
7.
Carcinogenesis ; 38(5): 492-503, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28334277

ABSTRACT

The highlight of the molecular basis and therapeutic targets of the bone-metastatic process requires the identification of biomarkers of metastasis colonization. Here, we studied miR-34a-5p expression, and Met-receptor expression and localization in bone metastases from ductal breast carcinomas, and in ductal carcinomas without history of metastasis (20 cases). miR-34a-5p was elevated in non-metastatic breast carcinoma, intermediate in the adjacent tissue and practically absent in bone metastases, opposite to pair-matched carcinoma. Met-receptor biomarker was highly expressed and inversely correlated with miR-34a-5p using the same set of bone-metastasis tissues. The miR-34a-5p silencing might depend on aberrant-epigenetic mechanisms of plastic-bone metastases, since in 1833 cells under methyltransferase blockade miR-34a-5p augmented. In fact, 1833 cells showed very low endogenous miR-34a-5p, in respect to parental MDA-MB231 breast carcinoma cells, and the restoration of miR-34a-5p with the mimic reduced Met and invasiveness. Notably, hepatocyte growth factor (HGF)-dependent Met stabilization was observed in bone-metastatic 1833 cells, consistent with Met co-distribution with the ligand HGF at plasma membrane and at nuclear levels in bone metastases. Met-protein level was higher in non-metastatic (low grade) than in metastatic (high grade) breast carcinomas, notwithstanding miR-34a-5p-elevated expression in both the specimens. Thus, mostly in non-metastatic carcinomas the elevated miR-34a-5p unaffected Met, important for invasive/mesenchymal phenotype, while possibly targeting some stemness biomarkers related to metastatic phenotype. In personalized therapies against bone metastasis, we suggest miR-34a-5p as a suitable target of epigenetic reprogramming leading to the accumulation of miR-34a-5p and the down-regulation of Met-tyrosine kinase, a key player of the bone-metastatic process.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Proto-Oncogene Proteins c-met/genetics , Biomarkers, Tumor , Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Survival/genetics , Disease Progression , Female , Hepatocyte Growth Factor/metabolism , Humans , Immunohistochemistry , Models, Biological , Proto-Oncogene Proteins c-met/metabolism
8.
Cell Death Dis ; 8(2): e2578, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28151481

ABSTRACT

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.


Subject(s)
Bone Neoplasms/genetics , Breast Neoplasms/genetics , Epigenesis, Genetic/genetics , Hepatocyte Growth Factor/genetics , Neoplasm Metastasis/genetics , Proto-Oncogene Proteins c-met/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Decitabine , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Suppressor Proteins/genetics
9.
Int J Mol Sci ; 18(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-28045433

ABSTRACT

Epigenetic mechanisms influence molecular patterns important for the bone-metastatic process, and here we highlight the role of WW-domain containing oxidoreductase (Wwox). The tumor-suppressor Wwox lacks in almost all cancer types; the variable expression in osteosarcomas is related to lung-metastasis formation, and exogenous Wwox destabilizes HIF-1α (subunit of Hypoxia inducible Factor-1, HIF-1) affecting aerobic glycolysis. Our recent studies show critical functions of Wwox present in 1833-osteotropic clone, in the corresponding xenograft model, and in human bone metastasis from breast carcinoma. In hypoxic-bone metastatic cells, Wwox enhances HIF-1α stabilization, phosphorylation, and nuclear translocation. Consistently, in bone-metastasis specimens Wwox localizes in cytosolic/perinuclear area, while TAZ (transcriptional co-activator with PDZ-binding motif) and HIF-1α co-localize in nuclei, playing specific regulatory mechanisms: TAZ is a co-factor of HIF-1, and Wwox regulates HIF-1 activity by controlling HIF-1α. In vitro, DNA methylation affects Wwox-protein synthesis; hypoxia decreases Wwox-protein level; hepatocyte growth factor (HGF) phosphorylates Wwox driving its nuclear shuttle, and counteracting a Twist program important for the epithelial phenotype and metastasis colonization. In agreement, in 1833-xenograft mice under DNA-methyltransferase blockade with decitabine, Wwox increases in nuclei/cytosol counteracting bone metastasis with prolongation of the survival. However, Wwox seems relevant for the autophagic process which sustains metastasis, enhancing more Beclin-1 than p62 protein levels, and p62 accumulates under decitabine consistent with adaptability of metastasis to therapy. In conclusion, Wwox methylation as a bone-metastasis therapeutic target would depend on autophagy conditions, and epigenetic mechanisms regulating Wwox may influence the phenotype of bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Bone and Bones/pathology , Breast Neoplasms/pathology , Breast/pathology , Epigenesis, Genetic , Oxidoreductases/genetics , Tumor Suppressor Proteins/genetics , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone and Bones/metabolism , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Female , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Oxidoreductases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , WW Domain-Containing Oxidoreductase
10.
Int J Mol Sci ; 17(5)2016 May 11.
Article in English | MEDLINE | ID: mdl-27187355

ABSTRACT

Bone metastatic cells release bone microenvironment proteins, such as the matricellular protein SPARC (secreted protein acidic and rich in cysteine), and share a cell signaling typical of the bone metabolism controlled by Runx2. The megakaryocytes in the bone marrow engrafted by the metastases seem to be one of the principal microenvironment sources of the biological stimuli, implicated in the formation of an osteoblastic niche, and affecting metastasis phenotype and colonization. Educated platelets in the circulation might derive from megakaryocytes in bone metastasis. The evaluation of predictive markers in the circulating platelets might be useful for the stratification of patients for therapeutic purposes. The hypoxic environment in bone metastasis is one of the key regulators of the network of the biological soluble and structural components of the matrix. In bone metastatic cells under hypoxia, similar patterns of Runx2 and SPARC are observed, both showing downregulation. Conversely, hypoxia induces Endothelin 1, which upregulates SPARC, and these biological stimuli may be considered prognostic markers of bone metastasis in breast carcinoma patients.


Subject(s)
Bone Neoplasms/metabolism , Oxygen/metabolism , Signal Transduction , Tumor Microenvironment , Animals , Bone Neoplasms/secondary , Cell Hypoxia , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
11.
Int J Mol Sci ; 17(4): 443, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-27023526

ABSTRACT

Autophagy favours metastatic growth through fuelling energy and nutrients and resistance to anoikis, typical of disseminated-tumour cells. The autophagic process, mediated by a unique organelle, the autophagosome, which fuses with lysosomes, is divided into three steps. Several stages, especially early omegasome formation and isolation-membrane initiation, remain controversial; molecular mechanisms involve the small-GTPase Rab5a, which regulates vesicle traffic for autophagosome formation. We examined Rab5a involvement in the function of key members of ubiquitin-conjugation systems, Atg7 and LC3-lipidated, interacting with the scaffold-protein p62. Immunohistochemistry of Rab5a was performed in human specimens of bone metastasis and pair-matched breast carcinoma; the autophagic-molecular mechanisms affected by Rab5a were evaluated in human 1833 bone metastatic cells, derived from breast-carcinoma MDA-MB231 cells. To clarify the role of Rab5a, 1833 cells were transfected transiently with Rab5a-dominant negative, and/or stably with the short-hairpin RNA Atg7, were exposed to two inhibitors of autolysosome function, and LC3II and p62 expression was measured. We showed basal autophagy in bone-metastatic cells and the pivotal role of Rab5a together with Beclin 1 between the early stages, elongation of isolation membrane/closed autophagosome mediated by Atg7, and the late-degradative stages. This regulatory network might occur in bone-metastasis and in high-grade dysplastic lesions, preceding invasive-breast carcinoma and conferring phenotypic characteristics for dissemination.


Subject(s)
Bone Neoplasms/pathology , Breast Neoplasms/pathology , rab5 GTP-Binding Proteins/metabolism , Autophagy , Autophagy-Related Protein 7/antagonists & inhibitors , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Neoplasm Staging , RNA Interference , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics
12.
Biochim Biophys Acta ; 1863(1): 64-76, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26481505

ABSTRACT

The pathogenesis of bone metastasis is unclear, and much focus in metastatic biology and therapy relays on epigenetic alterations. Since DNA-methyltransferase blockade with 5-aza-2'-deoxycytidine (dAza) counteracts tumour growth, here we utilized dAza to clarify whether molecular events undergoing epigenetic control were critical for bone metastatization. In particular, we investigated the patterns of secreted-protein acidic and rich in cysteine (SPARC) and of Endothelin 1, affected by DNA methyltransferases in tumours, with the hypothesis that in bone metastasis a coordinate function of SPARC and Endothelin 1, if any occurs, was orchestrated by DNA methylation. To this purpose, we prepared a xenograft model with the clone 1833, derived from human-MDA-MB231 cells, and dAza administration slowed-down metastasis outgrowth. This seemed consequent to the reductions of SPARC and Endothelin 1 at invasive front and in the bone marrow, mostly due to loss of Twist. In the metastasis bulk Snail, partly reduced by dAza, might sustain Endothelin 1-SPARC cooperativity. Both SPARC and Endothelin 1 underwent post-translational control by miRNAs, a molecular mechanism that might explain the in vivo data. Ectopic miR29a reduced SPARC expression also under long-term dAza exposure, while Endothelin 1 down-regulation occurred in the presence of endogenous-miR98 expression. Notably, dAza effects differed depending on in vivo and in vitro conditions. In 1833 cells exposed to 30-days dAza, SPARC-protein level was practically unaffected, while Endothelin 1 induction depended on the 3'-UTR functionality. The blockade of methyltransferases leading to SPARC reduction in vivo, might represent a promising strategy to hamper early steps of the metastatic process affecting the osteogenic niche.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , DNA Methylation , DNA, Neoplasm/metabolism , Tumor Microenvironment , Animals , Azacitidine/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA, Neoplasm/genetics , Endothelin-1/biosynthesis , Endothelin-1/genetics , Female , Humans , Mice , Mice, Nude , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Osteonectin/biosynthesis , Osteonectin/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics
13.
Int J Mol Sci ; 16(12): 28108-22, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26703564

ABSTRACT

In order to become established in the skeleton, metastatic cells disseminating from the breast carcinoma need to acquire organ-specific traits. There are no effective predictors for who will develop bone metastasis to guide long-term predictive therapy. Our purpose was to individuate events critical for bone colonization to make a molecular classification of breast carcinoma useful for bone-metastasis outcome. In dysplasia adjacent to carcinoma and in pair-matched specimens of bone metastasis we examined SPARC expression and localization as well as Endothelin 1/ETAR signals by immunohistochemistry, and the evaluation of plasma levels of SPARC by ELISA was also performed. In patients with breast carcinoma metastasizing to bone, SPARC and Endothelin 1/ETAR axis were highly expressed from dysplasia until bone metastasis, but the SPARC plasma level was as low as that of normal women, in contrast to patients that never develop bone metastasis, suggesting that circulating SPARC was counter adhesive. Altogether, the early identification of SPARC/Endothelin 1/ETAR in dysplastic lesions would be important to devise therapies preventing metastasis engraftment, since often carcinoma cells spread to distant organs at the time or even before patients present with cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/diagnosis , Breast Neoplasms/blood , Carcinoma/blood , Osteonectin/metabolism , Adult , Biomarkers, Tumor/blood , Bone Neoplasms/blood , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/metabolism , Carcinoma/pathology , Case-Control Studies , Endothelin-1/metabolism , Female , Humans , Middle Aged , Osteonectin/blood , Receptor, Endothelin A/metabolism
14.
Mol Cancer ; 14: 112, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26041563

ABSTRACT

BACKGROUND: Much effort has been devoted to determining how metastatic cells and microenvironment reciprocally interact. However, the role of biological stimuli of microenvironment in controlling molecular events in bone metastasis from breast carcinoma for mesenchymal-epithelial transition (MET) is largely unknown. The purpose of the present paper was to clarify (1) the influence of hepatocyte-growth factor (HGF) and transforming growth factorß1 (TGFß1) on the phenotype of bone-metastatic 1833 and parental MDA-MB231 cells; (2) the hierarchic response of Twist and Snail controlled by Wwox co-factor, that might be critical for the control of 1833-adhesive properties via E-cadherin. METHODS: We studied under HGF and TGFß1 the gene profiles-responsible for epithelial-mesenchymal transition (EMT), versus the revertant MET phenotype-making the correspondence with 1833 morphology and the relation to HGF-dependent control of TGFß1 signalling. In particular, the activation of Twist program and the underlying molecular mechanisms were investigated, considering the role of endogenous and exogenous Wwox with siRNAWWOX and the expression vector transfection, to clarify whether Twist affected E-cadherin transactivation through a network of transcription factors and regulators. RESULTS: HGF and TGFß1 oppositely affected the expression of Wwox in 1833 cells. Under HGF, endogenous Wwox decreased concomitant with Twist access to nuclei and its phosphorylation via PI3K/Akt pathway. Twist activated by HGF did not influence the gene profile through an E-box mechanism, but participated in the interplay of PPARγ/Ets1/NF-kB-transcription factors, triggering E-cadherin transactivation. Altogether, HGF conferred MET phenotype to 1833 cells, even if this was transient since followed by TGFß1-signalling activation. TGFß1 induced Snail in both the cell lines, with E-cadherin down-regulation only in 1833 cells because in MDA-MB231 cells E-cadherin was practically absent. Exogenous Wwox activated metastatic HIF-1, with Twist as co-factor. CONCLUSIONS: HGF and TGFß1 of bone-metastasis microenvironment acted co-ordinately, influencing non redundant pathways regulated by Twist program or Snail-transcription factor, with reversible MET switch. This process implicated different roles for Wwox in the various steps of the metastatic process including colonization, with microenvironmental/exogenous Wwox that activated HIF-1, important for E-cadherin expression. Interfering with the Twist program by targeting the pre-metastatic niche stimuli could be an effective anti-bone metastasis therapy.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Hepatocyte Growth Factor/metabolism , Nuclear Proteins/metabolism , Oxidoreductases/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Suppressor Proteins/metabolism , Twist-Related Protein 1/metabolism , Animals , Bone Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , DNA, Neoplasm/metabolism , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1/metabolism , Mice, Nude , Models, Biological , Phosphorylation , Protein Binding , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/metabolism , Transcriptional Activation/genetics , WW Domain-Containing Oxidoreductase
15.
Exp Cell Res ; 330(2): 287-299, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25447306

ABSTRACT

The present study deals with the molecular mechanisms involved in the regulation of E-cadherin expression under hypoxia, because the adjustment of the amount of E-cadherin due to physical stimuli of the microenvironment might influence the colonization of metastasis to skeleton. We analyzed the effect of 1% oxygen tension, that is similar to that encountered in the bone marrow by metastatic cells spreading from breast carcinoma. The purpose was to evaluate the hypoxia-orchestrated control of E-cadherin transactivation via hypoxia inducible factor-1 (HIF-1) and peroxisome proliferator activated receptor-γ (PPARγ), and the involvement of Hippo pathway members, as regulators of transcription factors. To give a translational significance to the study, we took into consideration human pair-matched ductal breast carcinoma and bone metastasis: E-cadherin and Wwox were expressed in bone metastasis but not in breast carcinoma, while HIF-1α and TAZ seemed localized principally in nuclei of metastasis and were found in all cell compartments of breast carcinoma. A close examination of the regulatory mechanisms underlying E-cadherin expression in bone metastasis was done in 1833 clone derived from MDA-MB231 cells. Hypoxia induced E-cadherin only in 1833 clone, but not in parental cells, through HIF-1 and PPARγ activities, while Wwox decreased. Since Wwox was highly expressed in bone metastasis, the effect of ectopic Wwox was evaluated, and we showed E-cadherin transactivation and enhanced invasiveness in WWOX transfected 1833 cells. Also, hypoxia was additive with ectopic Wwox remarkably enhancing HIF-1α nuclear shuttle and accumulation due to the lengthening of the half-life of HIF-1α protein; under this experimental condition HIF-1α appeared as a slower migrated band compared with control, in agreement with the phosphorylation state. The in vitro data strongly supported the almost exclusive presence of HIF-1α in nuclei of human-bone metastasis. Thus, we identified Wwox as a novel molecule in the HIF-1α-HDM2 regulatory loop, necessary for the dynamic regulation of the HIF-1α amount, and we suggested that the reduction of endogenous Wwox free pool under hypoxia might also be due to the interaction with HDM2, sequestering the E3 ubiquitin ligase. We highlighted the importance of nuclear HIF-1α in the biology of metastasis for the mesenchymal-epithelial transition: this phenotype was regulated by Wwox plus hypoxia through E-cadherin target gene, playing a pivotal role in bone metastasis colonization.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cadherins/genetics , Carcinoma, Ductal, Breast/secondary , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oxidoreductases/genetics , Tumor Suppressor Proteins/genetics , Acyltransferases , Cadherins/biosynthesis , Cadherins/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/biosynthesis , PPAR gamma/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcriptional Activation , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/biosynthesis , Ubiquitin-Protein Ligases/metabolism , WW Domain-Containing Oxidoreductase
16.
Biochim Biophys Acta ; 1843(4): 815-26, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24373848

ABSTRACT

The present study was undertaken to clarify the function(s) of Endothelin-1 and its receptors ETAR and ETBR in osteolytic-bone metastasis from breast cancer, and their regulation by hepatocyte and transforming growth factors (HGF, TGF-ß) and hypoxia. The aim was to evaluate the adaptability of bone metastasis to microenvironmental stimuli through Endothelin-1-mediated epithelial-mesenchymal transition (EMT), or the reverse process MET, and through osteomimicry possible key features for bone colonization. We compared low (MCF-7) and high (MDA-MB231) invasive-breast carcinoma cells, and 1833-bone metastatic clone, with human pair-matched primary breast-carcinomas and bone metastases. Parental MDA-MB231 and the derived 1833-clone responded oppositely to the stimuli. In 1833 cells, TGF-ß and hypoxia increased Endothelin-1 release, altogether reducing invasiveness important for engraftment, while Endothelin-1 enhanced MDA-MB231 cell invasiveness. The Endothelin-1-autocrine loop contributed to the cooperation of intracellular-signaling pathways and extracellular stimuli triggering MET in 1833 cells, and EMT in MDA-MB231 cells. Only in 1833 cells, HGF negatively influenced transactivation and release of Endothelin-1, suggesting a temporal sequence of these stimuli with an initial role of HGF-triggered Wnt/ß-catenin pathway in metastatization. Then, Endothelin-1/ETAR conferred MET and osteomimetic phenotypes, with Runt-related transcription factor 2 activation and metalloproteinase 9 expression, contributing to colonization and osteolysis. Findings with human pair-matched primary ductal carcinomas and bone metastases gave a translational significance to the molecular study. Endothelin-1, ETAR and ETBR correlated with the acquisition of malignant potential, because of high expression already in the in situ carcinoma. These molecular markers might be used as predictive index of aggressive behavior and invasive/metastatic phenotype.


Subject(s)
Bone Neoplasms/genetics , Breast Neoplasms/genetics , Endothelin-1/genetics , Epithelial-Mesenchymal Transition/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Endothelin-1/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Invasiveness/genetics , Receptor, Endothelin A , Receptor, Endothelin B , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Wnt Signaling Pathway/genetics
17.
Exp Cell Res ; 319(12): 1865-1874, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23660387

ABSTRACT

Here, we report a complex regulation of endothelin-1 (ET-1) axis driven by epigenetic reactions in 1833-bone metastatic cells, emphasizing the importance in skeletal metastasis from breast carcinoma. Inhibitors of histone deacetylases, trichostatin A (TSA), and of DNA methylases, 5'-Azacytidine (Aza), caused, respectively, reduction and increase in 1833 cell invasiveness, without affecting the basal migration of parental MDA-MB231 cells. Of note, in the two cell lines exposed to Aza the blockade of the ET-1 receptor ETAR with BQ-123 oppositely changed invasive properties. Even if in MDA-MB231 cells the ET-1 axis was scarcely influenced by epigenetic reactions, ETAR remarkably decreased after Aza. In contrast, in 1833 cells Aza exposure enhanced ET-1 coupled to ETAR wild type, being also ETAR truncated form increased, and invasiveness was stimulated. Under demethylation, the increase in ET-1 steady state protein level in 1833 clone seemed regulated at transcriptional level principally via Ets1 transcription factor. In fact, actinomycin D almost completely prevented ET-1 mRNA induction due to Aza. Only in 1833 cells, TSA exposure inactivated ET-1 axis, with reduction of the expression of ET-1 and ETAR mutated form, in agreement with Matrigel invasion decrease. This treatment favoured the ET-1 repressional control, taking place at the level of mRNA stability due to the 3'-untranslated region in the ET-1 gene, and also decreased transcription via NF-kB. Environmental conditions that alter the balance between epigenetic reactions might, therefore, affect metastasis migratory mode influencing ET-1 axis.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/genetics , Carcinoma/genetics , Cell Movement , Endothelin-1/genetics , Epigenesis, Genetic , Azacitidine/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/metabolism , Carcinoma/secondary , Cell Line, Tumor , Collagen/metabolism , DNA Methylation , Dactinomycin/pharmacology , Drug Combinations , Endothelin A Receptor Antagonists , Endothelin-1/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Laminin/metabolism , Mutation , Neoplasm Invasiveness/genetics , Peptides, Cyclic/pharmacology , Proteoglycans/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , RNA Stability , RNA, Messenger/metabolism , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism , Transcription, Genetic
18.
Eur J Cancer ; 49(11): 2608-18, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23566416

ABSTRACT

The hypoxic microenvironment of bone marrow favours the bone metastasis process. Hypoxia inducible factor (HIF)-1α is hallmark for hypoxia, correlating with poor prognosis and radio/chemotherapy resistance of primary-breast carcinoma. For bone metastasis, the molecular mechanisms involved in HIF-1α expression and HIF-1 (α/ß heterodimer)-transcription factor activity are scarcely known. We studied the role played by HIF-1 in the cross-talk between neoplastic and supportive-microenvironmental cells. Also, WWdomain-containing oxidoreductase (Wwox) and transcriptional co-activator with PDZ-binding motif (TAZ) were taken into consideration evaluating whether these Hippo-pathway effectors affect bone-metastatic phenotype through HIF-1 activity. Considering bone-metastasis specimens, nuclear HIF-1α-TAZ co-localisation occurred in neoplastic and supportive cells, such as fibroblasts and endotheliocytes. Based on these data, the functional importance was verified using 1833-bone metastatic clone under hypoxia: nuclear HIF-1α and TAZ expression increased and co-immunoprecipitated, activating HIF-1-DNA binding and transactivation. In contrast, Wwox localised at perinuclear level in neoplastic cells of bone metastasis, being almost absent in supportive cells, and Wwox-protein expression diminished in hypoxic-1833 cells. Thus, TAZ regulation of HIF-1 activity might be important for bone-secondary growth, participating in metastasis-stroma cross-talk. Further, TAZ and HIF-1α-protein levels seemed correlated. In fact, blocking cyclooxygenase-2 with NS398 in hypoxic-1833 cells, not only HIF-1α decreased but also molecular-mechanism(s) upstream of the Hippo pathway were triggered: LATS-dependent TAZ phosphorylation seemed responsible for TAZ nucleus/cytoplasm translocation and degradation. In the 1833-xenograft model, NS398 largely prevented the outgrowth of bone-metastatic cells, probably related to remarkable-extracellular matrix assembly. We gained clinical insight into HIF-1α and TAZ as candidate biomarkers for bone avidity, relevant for early-therapeutic intervention against bone metastasis.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Hypoxia/physiology , Hypoxia-Inducible Factor 1/metabolism , Oxidoreductases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Acyltransferases , Bone Neoplasms/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Hypoxia/genetics , Female , Humans , Hypoxia-Inducible Factor 1/genetics , Immunohistochemistry , PDZ Domains , Transcription Factors/genetics , Transfection , Tumor Microenvironment , WW Domain-Containing Oxidoreductase
19.
Eur J Cancer ; 49(1): 231-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22717556

ABSTRACT

We investigated the involvement of Hippo-related pathways in bone metastasis from breast cancer, by evaluating E-cadherin expression downstream of WWdomain-containing oxidoreductase (Wwox) and transcriptional co-activator with PDZ-binding motif (TAZ). These nuclear effectors functioned in a context-specific fashion on transcriptome, depending on breast-cancer aggressiveness and methylation state. Wwox and E-cadherin were found in human specimens of bone metastasis but not in primary-ductal breast carcinoma, while TAZ showed a characteristic localisation in metastasis nuclei. Wwox and E-cadherin were higher in 1833-metastatic clone with bone avidity than in parental-MDA-MB231 cells, while only metastatic cells presented TAZ. In 1833 cells, a complex interplay of transcriptional signalling controlled E-cadherin transactivation. Wwox and TAZ activated Hypoxia inducible factor-1 (HIF-1) binding to E-cadherin promoter, while Peroxisome proliferator-activated receptor γ (PPARγ) intervened in E-cadherin transactivation favouring and preventing Wwox and TAZ functions, respectively. Methylation impinged on Hippo-related pathways through Wwox and TAZ, modifying metastatic phenotype. The protract exposure to 5-azacytidine (Aza), by affecting methylation state modified the shape of 1833 cells, becoming mesenchymal as that of MDA-MB231 cells and reduced spontaneous-Matrigel invasion. The underlying-molecular mechanisms were diminutions of E-cadherin, Wwox, matrix metalloproteases 2 and 9, HIF-1- and PPARγ-activities, inversely correlated to Snail and nuclear-TAZ accumulations. Exogenous WWOX restored 1833-Aza invasion. Thus, 1833-Aza cells permitted to study the role played by methylation in metastasis plasticity, being E-cadherin loss part of an entire-gene reprogramming. Of note, bone-metastasis formation in 1833-Aza xenograft was partially impaired, prolonging mice survival. In conclusion, the methylation-heritable changes seemed important for cancer progression to establish bone metastasis engraftment/growth, by affecting steps requiring homotipic and/or heterotypic-adhesive properties and matrix degradation.


Subject(s)
Bone Neoplasms/genetics , Breast Neoplasms/genetics , Cadherins/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Metastasis/genetics , Oxidoreductases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , DNA Methylation , Electrophoretic Mobility Shift Assay , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Neoplasm Metastasis/pathology , Oxidoreductases/genetics , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transcriptome , Tumor Suppressor Proteins/genetics , WW Domain-Containing Oxidoreductase
20.
Biochem Biophys Res Commun ; 428(2): 271-7, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23085045

ABSTRACT

The human adipose-tissue derived stem/stromal cells (hASCs) are an interesting source for bone-tissue engineering applications. Our aim was to clarify in hASCs the role of acetylation in the control of Runt-related transcription factor 2 (Runx2) and Peroxisome proliferator activated receptor (PPAR) γ. These key osteogenic and adipogenic transcription factors are oppositely involved in osteo-differentiation. The hASCs, committed or not towards bone lineage with osteoinductive medium, were exposed to HDACs chemical blockade with Trichostatin A (TSA) or were genetically silenced for HDACs. Alkaline phosphatase (ALP) and collagen/calcium deposition, considered as early and late osteogenic markers, were evaluated concomitantly as index of osteo-differentiation. TSA pretreatment, useful experimental protocol to analyse pan-HDAC-chemical inhibition, and switch to osteogenic medium induced early-osteoblast maturation gene Runx2, while transiently decreased PPARγ and scarcely affected late-differentiation markers. Time-dependent effects were observed after knocking-down of HDAC1 and 3: Runx2 and ALP underwent early activation, followed by late-osteogenic markers increase and by PPARγ/ALP activity diminutions mostly after HDAC3 silencing. HDAC1 and 3 genetic blockade increased and decreased Runx2 and PPARγ target genes, respectively. Noteworthy, HDACs knocking-down favoured the commitment effect of osteogenic medium. Our results reveal a role for HDACs in orchestrating osteo-differentiation of hASCs at transcriptional level, and might provide new insights into the modulation of hASCs-based regenerative therapy.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/physiology , Histone Deacetylases/physiology , Osteogenesis/physiology , Stem Cells/cytology , Adipogenesis/drug effects , Adipogenesis/genetics , Adipogenesis/physiology , Adipose Tissue/drug effects , Adult , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Gene Knockdown Techniques , Histone Deacetylase 1/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Hydroxamic Acids/pharmacology , Middle Aged , Osteogenesis/drug effects , Osteogenesis/genetics , PPAR gamma/metabolism , Regenerative Medicine , Stem Cells/drug effects , Suppression, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...