Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 9(12): e114530, 2014.
Article in English | MEDLINE | ID: mdl-25485970

ABSTRACT

The reciprocal interaction between cancer cells and the tissue-specific stroma is critical for primary and metastatic tumor growth progression. Prostate cancer cells colonize preferentially bone (osteotropism), where they alter the physiological balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption, and elicit prevalently an osteoblastic response (osteoinduction). The molecular cues provided by osteoblasts for the survival and growth of bone metastatic prostate cancer cells are largely unknown. We exploited the sufficient divergence between human and mouse RNA sequences together with redefinition of highly species-specific gene arrays by computer-aided and experimental exclusion of cross-hybridizing oligonucleotide probes. This strategy allowed the dissection of the stroma (mouse) from the cancer cell (human) transcriptome in bone metastasis xenograft models of human osteoinductive prostate cancer cells (VCaP and C4-2B). As a result, we generated the osteoblastic bone metastasis-associated stroma transcriptome (OB-BMST). Subtraction of genes shared by inflammation, wound healing and desmoplastic responses, and by the tissue type-independent stroma responses to a variety of non-osteotropic and osteotropic primary cancers generated a curated gene signature ("Core" OB-BMST) putatively representing the bone marrow/bone-specific stroma response to prostate cancer-induced, osteoblastic bone metastasis. The expression pattern of three representative Core OB-BMST genes (PTN, EPHA3 and FSCN1) seems to confirm the bone specificity of this response. A robust induction of genes involved in osteogenesis and angiogenesis dominates both the OB-BMST and Core OB-BMST. This translates in an amplification of hematopoietic and, remarkably, prostate epithelial stem cell niche components that may function as a self-reinforcing bone metastatic niche providing a growth support specific for osteoinductive prostate cancer cells. The induction of this combinatorial stem cell niche is a novel mechanism that may also explain cancer cell osteotropism and local interference with hematopoiesis (myelophthisis). Accordingly, these stem cell niche components may represent innovative therapeutic targets and/or serum biomarkers in osteoblastic bone metastasis.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/secondary , Epithelial Cells/pathology , Hematopoietic System/pathology , Osteoblasts/pathology , Prostatic Neoplasms/pathology , Stem Cell Niche/genetics , Stromal Cells/pathology , Animals , Biomarkers, Tumor/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Differentiation , Epithelial Cells/metabolism , Gene Expression Profiling , Hematopoietic System/metabolism , Humans , Immunoenzyme Techniques , Male , Mice , Neovascularization, Pathologic/genetics , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Osteogenesis/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Tumor Cells, Cultured
2.
PLoS One ; 6(1): e16078, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21249149

ABSTRACT

Members of the BMP and Wnt protein families play a relevant role in physiologic and pathologic bone turnover. Extracellular antagonists are crucial for the modulation of their activity. Lack of expression of the BMP antagonist noggin by osteoinductive, carcinoma-derived cell lines is a determinant of the osteoblast response induced by their bone metastases. In contrast, osteolytic, carcinoma-derived cell lines express noggin constitutively. We hypothesized that cancer cell-derived noggin may contribute to the pathogenesis of osteolytic bone metastasis of solid cancers by repressing bone formation. Intra-osseous xenografts of PC-3 prostate cancer cells induced osteolytic lesions characterized not only by enhanced osteoclast-mediated bone resorption, but also by decreased osteoblast-mediated bone formation. Therefore, in this model, uncoupling of the bone remodeling process contributes to osteolysis. Bone formation was preserved in the osteolytic lesions induced by noggin-silenced PC-3 cells, suggesting that cancer cell-derived noggin interferes with physiologic bone coupling. Furthermore, intra-osseous tumor growth of noggin-silenced PC-3 cells was limited, most probably as a result of the persisting osteoblast activity. This investigation provides new evidence for a model of osteolytic bone metastasis where constitutive secretion of noggin by cancer cells mediates inhibition of bone formation, thereby preventing repair of osteolytic lesions generated by an excess of osteoclast-mediated bone resorption. Therefore, noggin suppression may be a novel strategy for the treatment of osteolytic bone metastases.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Neoplasms/etiology , Carrier Proteins/physiology , Osteolysis/etiology , Prostatic Neoplasms/pathology , Signal Transduction , Bone Neoplasms/secondary , Bone Remodeling , Bone Resorption , Cell Line, Tumor , Humans , Male , Osteoblasts/pathology
3.
Prostate ; 68(12): 1307-18, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18512728

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the inhibitory growth effects of different potential chemopreventive agents in vitro and to determine their influence on PSA mRNA and protein expression with an established screening platform. METHODS: LNCaP and C4-2 cells were incubated with genistein, seleno-L-methionine, lycopene, DL-alpha-tocopherol, and trans-beta-carotene at three different concentrations and cell growth was determined by the MTT assay. PSA mRNA expression was assessed by quantitative real-time RT-PCR and secreted PSA protein levels were quantified by the microparticle enzyme immunoassay. RESULTS: Genistein, seleno-l-methionine and lycopene inhibited LNCaP cell growth, and the proliferation of C4-2 cells was suppressed by seleno-L-methionine and lycopene. PSA mRNA expression was downregulated by genistein in LNCaP but not C4-2 cells. No other compound tested altered PSA mRNA expression. PSA protein expression was downregulated by genistein, seleno-L-methionine, DL-alpha-tocopherol in LNCaP cells. In C4-2 cells only genistein significantly reduced the secretion of PSA protein. CONCLUSIONS: In the LNCaP progression model PSA expression depends on the compound, its concentration and on the hormonal dependence of the cell line used and does not necessarily reflect cell growth or death. Before potential substances are evaluated in clinical trials using PSA as a surrogate end point marker, their effect on PSA mRNA and protein expression has to be considered to correctly assess treatment response by PSA.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Anticarcinogenic Agents/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Adenocarcinoma/prevention & control , Biomarkers, Tumor/metabolism , Carotenoids/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Genistein/pharmacology , Humans , Lycopene , Male , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/prevention & control , RNA, Messenger/metabolism , Selenomethionine/pharmacology , Vitamins/pharmacology , alpha-Tocopherol/pharmacology , beta Carotene/pharmacology
4.
Cancer Res ; 67(18): 8742-51, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875715

ABSTRACT

Bone morphogenetic protein 7 (BMP7) counteracts the physiological epithelial-to-mesenchymal transition (EMT), a process that is indicative of epithelial plasticity. Because EMT is involved in cancer, we investigated whether BMP7 plays a role in breast cancer growth and metastasis. In this study, we show that decreased BMP7 expression in primary breast cancer is significantly associated with the formation of clinically overt bone metastases in patients with > or = 10 years of follow-up. In line with these clinical observations, BMP7 expression is inversely related to tumorigenicity and invasive behavior of human breast cancer cell lines. Moreover, BMP7 decreased the expression of vimentin, a mesenchymal marker associated with invasiveness and poor prognosis, in human MDA-MB-231 (MDA-231)-B/Luc(+) breast cancer cells under basal and transforming growth factor-beta (TGF-beta)-stimulated conditions. In addition, exogenous addition of BMP7 to TGF-beta-stimulated MDA-231 cells inhibited Smad-mediated TGF-beta signaling. Furthermore, in a well-established bone metastasis model using whole-body bioluminescent reporter imaging, stable overexpression of BMP7 in MDA-231 cells inhibited de novo formation and progression of osteolytic bone metastases and, hence, their metastatic capability. In line with these observations, daily i.v. administration of BMP7 (100 mug/kg/d) significantly inhibited orthotopic and intrabone growth of MDA-231-B/Luc(+) cells in nude mice. Our data suggest that decreased BMP7 expression during carcinogenesis in the human breast contributes to the acquisition of a bone metastatic phenotype. Because exogenous BMP7 can still counteract the breast cancer growth at the primary site and in bone, BMP7 may represent a novel therapeutic molecule for repression of local and bone metastatic growth of breast cancer.


Subject(s)
Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Progression , Epithelial Cells/pathology , Female , Humans , Mesoderm/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Retrospective Studies , Signal Transduction , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
5.
Am J Pathol ; 171(3): 1047-57, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17724140

ABSTRACT

Bone morphogenic protein 7 (BMP7) counteracts physiological epithelial-to-mesenchymal transition, a process that is indicative of epithelial plasticity. Because epithelial-to-mesenchymal transition is involved in cancer, we investigated whether BMP7 plays a role in prostate cancer growth and metastasis. BMP7 expression in laser-microdissected primary human prostate cancer tissue was strongly down-regulated compared with normal prostate luminal epithelium. Furthermore, BMP7 expression in prostate cancer cell lines was inversely related to tumorigenic and metastatic potential in vivo and significantly correlated to E-cadherin/vimentin ratios. Exogenous addition of BMP7 to human prostate cancer cells dose-dependently inhibited transforming growth factor beta-induced activation of nuclear Smad3/4 complexes via ALK5 and induced E-cadherin expression. Moreover, BMP7-induced activation of nuclear Smad1/4/5 signaling transduced via BMP type I receptors was synergistically stimulated in the presence of transforming growth factor beta, a growth factor that is enriched in the bone microenvironment. Daily BMP7 administration to nude mice inhibited the growth of cancer cells in bone. In contrast, no significant growth inhibitory effect of BMP7 was observed in intraprostatic xenografts. Collectively, our observations suggest that BMP7 controls and preserves the epithelial phenotype in the human prostate and underscore a decisive role of the tumor microenvironment in mediating the therapeutic response of BMP7. Thus, BMP7 can still counteract the epithelial-to-mesenchymal transition process in the metastatic tumor, positioning BMP7 as a novel therapeutic molecule for treatment of metastatic bone disease.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Bone Neoplasms/secondary , Epithelial Cells/physiology , Homeostasis , Prostate , Prostatic Neoplasms , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Cell Line, Tumor , Epithelial Cells/cytology , Humans , Male , Mesoderm/cytology , Mesoderm/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Prostate/cytology , Prostate/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism
6.
Am J Pathol ; 170(1): 160-75, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200191

ABSTRACT

Prostate and mammary cancer bone metastases can be osteoblastic or osteolytic, but the mechanisms determining these features are unclear. Bone morphogenetic and Wnt proteins are osteoinductive molecules. Their activity is modulated by antagonists such as noggin and dickkopf-1. Differential expression analysis of bone morphogenetic and Wnt protein antagonists in human prostate and mammary cancer cell lines showed that osteolytic cell lines constitutively express in vitro noggin and dickkopf-1 and at least one of the osteolytic cytokines parathyroid hormone-related protein, colony-stimulating factor-1, and interleukin-8. In contrast, osteoinductive cell lines express neither noggin nor dickkopf-1 nor osteolytic cytokines in vitro. The noggin differential expression profile observed in vitro was confirmed in vivo in prostate cancer cell lines xenografted into bone and in clinical samples of bone metastasis. Forced noggin expression in an osteoinductive prostate cancer cell line abolished the osteoblast response induced in vivo by its intraosseous xenografts. Basal bone resorption and tumor growth kinetics were marginally affected. Lack of noggin and possibly dickkopf-1 expression by cancer cells may be a relevant mechanism contributing to the osteoblast response in bone metastases. Concomitant lack of osteolytic cytokines may be permissive of this effect. Noggin is a candidate drug for the adjuvant therapy of bone metastasis.


Subject(s)
Bone Neoplasms , Carrier Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Osteoblasts , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Differentiation/genetics , Cell Proliferation , Cytokines/biosynthesis , Female , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Transplantation , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
7.
Int J Cancer ; 118(4): 899-906, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16106424

ABSTRACT

The transforming growth factor-beta (TGFbeta) superfamily and its downstream effector genes are key regulators of epithelial homeostasis. Altered expression of these genes may be associated with malignant transformation of the prostate gland. The cDNA array analysis of differential expression of the TGFbeta superfamily and functionally related genes between patient-matched noncancerous prostate (NP) and prostate cancer (PC) bulk tissue specimens highlighted two genes, namely TGFbeta-stimulated clone-22 (TSC-22) and Id4. Verification of their mRNA expression by real-time PCR in patient-matched NP and PC bulk tissue, in laser-captured pure epithelial and cancer cells and in NP and PC cell lines confirmed TSC-22 underexpression, but not Id4 overexpression, in PC and in human PC cell lines. Immunohistochemical analysis showed that TSC-22 protein expression in NP is restricted to the basal cells and colocalizes with the basal cell marker cytokeratin 5. In contrast, all matched PC samples lack TSC-22 immunoreactivity. Likewise, PC cell lines do not show detectable TSC-22 protein expression as shown by immunoblotting. TSC-22 should be considered as a novel basal cell marker, potentially useful for studying lineage determination within the epithelial compartment of the prostate. Conversely, lack of TSC-22 seems to be a hallmark of malignant transformation of the prostate epithelium. Accordingly, TSC-22 immunohistochemistry may prove to be a diagnostic tool for discriminating benign lesions from malignant ones of the prostate. The suggested tumour suppressor function of TSC-22 warrants further investigation on its role in prostate carcinogenesis and on the TSC-22 pathway as a candidate therapeutic target in PC.


Subject(s)
Inhibitor of Differentiation Proteins/biosynthesis , Prostate/physiology , Prostatic Neoplasms/genetics , Repressor Proteins/biosynthesis , Transforming Growth Factor beta/physiology , Aged , Cell Transformation, Neoplastic , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Prostate/cytology , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , Tumor Cells, Cultured
8.
Cancer Immunol Immunother ; 53(11): 1005-17, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15185010

ABSTRACT

HER-2/neu, a tumor-associated antigen (TAAg), plays a critical role in oncogenesis of various tumor types, and its selective overexpression by malignant tumor cells makes it an ideal target for immunotherapy. A prerequisite for clinical vaccines is the construction of safe and highly immunogenic reagents able to generate efficient immune responses against TAAg. Previous protein vaccines, consisting of the extracellular domain of HER-2/neu (pNeuECD), were shown to elicit an immune response that did not provide protection from transplantable tumors expressing HER-2/neu. Here we showed that virosomes, which consist of reconstituted viral envelopes without viral genetic material, can act as a carrier and an adjuvant for a truncated protein pNeuECD. Mice vaccinated with pNeuECD either encapsulated in virosomes or bound to the virosomal membrane (Vir-pNeuECD), generated rNeu-specific humoral and cytotoxic immune responses. In addition, Vir-p(NeuECD) induced significant tumor rejection and additionally did not lead to delayed tumor formation when compared with free pNeuECD in complete Freund's adjuvant. There was no difference between the virosomal constructs. Taken together these results suggest that virosomes, as clinically approved safe vaccines, can be used to elicit both humoral and cell-mediated responses against TAAg and induce tumor rejection. Our model is providing important preclinical data to design human vaccination trials for patients with tumors overexpressing HER-2/neu, either as a primary vaccination or as a boost in combination with other vaccines in a context of an adjuvant treatment plan.


Subject(s)
Cancer Vaccines , Immunotherapy/methods , Receptor, ErbB-2/chemistry , Virosomes/chemistry , Animals , Blotting, Western , COS Cells , Cell Separation , Cloning, Molecular , DNA, Complementary/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/metabolism , Flow Cytometry , Freund's Adjuvant , Genetic Vectors , Heterozygote , Humans , Immunity, Cellular , Mice , Mice, Transgenic , Plasmids/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Rats , Time Factors , Transfection , Vaccinia virus/genetics , Viral Envelope Proteins/chemistry
9.
Cancer Res ; 62(2): 437-44, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11809693

ABSTRACT

HER-2/neu (p185(HER2)) oncogene represents an attractive target for antibody-mediated immunotherapy. The major problem of combining chemotherapy and immunotherapy is the severe side effects that limit the use of doxorubicin (Doxo) as a cytotoxic drug. We have used virosomes (Vir; reconstituted fusion-active viral envelopes) as a new drug delivery system and have shown that Vir are capable of binding and penetrating into tumor cells, delivering cytotoxic drugs. We have additionally demonstrated that conjugating Fab' fragments of an antirat Neu (anti-rNeu) monoclonal antibody to Vir selectively and efficiently inhibits tumor progression of established rNeu-overexpressing breast tumors. Fab'-Doxo-Vir combine the antiproliferative properties of the monoclonal antibody and the cytotoxic effect of Doxo in vivo. Furthermore, Fab'-Doxo-Vir significantly inhibit tumor formation at a tumor load representing metastatic spread. These results indicate that Vir conjugated with an antibody against a tumor antigen are a promising new selective drug delivery system for the treatment of tumors expressing a specific tumor antigen.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Immunoglobulin Fragments/administration & dosage , Immunotoxins/administration & dosage , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/toxicity , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Doxorubicin/toxicity , Drug Delivery Systems , Immunoglobulin Fragments/metabolism , Immunoglobulin Fragments/toxicity , Immunotoxins/pharmacokinetics , Immunotoxins/toxicity , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Virosomes
SELECTION OF CITATIONS
SEARCH DETAIL
...