Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Lab Invest ; 100(3): 400-413, 2020 03.
Article in English | MEDLINE | ID: mdl-31570773

ABSTRACT

TBX3 is a member of the highly conserved family of T-box transcription factors involved in embryogenesis, organogenesis and tumor progression. While the functional role of TBX3 in tumorigenesis has been widely studied, less is known about the specific functions of the different isoforms (TBX3iso1 and TBX3iso2) which differ in their DNA-binding domain. We therefore sought to investigate the functional consequence of this highly conserved splice event as it relates to TBX3-induced tumorigenesis. By utilizing a nude mouse xenograft model, we have identified differential tumorigenic potential between TBX3 isoforms, with TBX3iso1 overexpression more commonly associated with invasive carcinoma and high tumor vascularity. Transcriptional analysis of signaling pathways altered by TBX3iso1 and TBX3iso2 overexpression revealed significant differences in angiogenesis-related genes. Importantly, osteopontin (OPN), a cancer-associated secreted phosphoprotein, was significantly up-regulated with TBX3iso1 (but not TBX3iso2) overexpression. This pattern was observed across three non/weakly-tumorigenic breast cancer cell lines (21PT, 21NT, and MCF7). Up-regulation of OPN in TBX3iso1 overexpressing cells was associated with induction of hyaluronan synthase 2 (HAS2) expression and increased retention of hyaluronan in pericellular matrices. These transcriptional changes were accompanied by the ability to induce endothelial cell vascular channel formation by conditioned media in vitro, which could be inhibited through addition of an OPN neutralizing antibody. Within the TCGA breast cancer cohort, we identified an 8.1-fold higher TBX3iso1 to TBX3iso2 transcript ratio in tumors relative to control, and this ratio was positively associated with high-tumor grade and an aggressive molecular subtype. Collectively, the described changes involving TBX3iso1-dependent promotion of angiogenesis may thus serve as an adaptive mechanism within breast cancer cells, potentially explaining differences in tumor formation rates between TBX3 isoforms in vivo. This study is the first of its kind to report significant functional differences between the two TBX3 isoforms, both in vitro and in vivo.


Subject(s)
Breast Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Protein Isoforms , T-Box Domain Proteins , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/pathology , Osteopontin/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
2.
J Pathol ; 248(2): 191-203, 2019 06.
Article in English | MEDLINE | ID: mdl-30697731

ABSTRACT

The acquisition of cellular invasiveness by breast epithelial cells and subsequent transition from ductal carcinoma in situ (DCIS) to invasive breast cancer is a critical step in breast cancer progression. Little is known about the molecular dynamics governing this transition. We have previously shown that overexpression of the transcriptional regulator TBX3 in DCIS-like cells increases survival, growth, and invasiveness. To explore this mechanism further and assess direct transcriptional targets of TBX3 in a high-resolution, isoform-specific context, we conducted genome-wide chromatin-immunoprecipitation (ChIP) arrays coupled with transcriptomic analysis. We show that TBX3 regulates several epithelial-mesenchymal transition (EMT)-related genes, including SLUG and TWIST1. Importantly, we demonstrate that TBX3 is a direct regulator of SLUG expression, and SLUG expression is required for TBX3-induced migration and invasion. Assessing TBX3 by immunohistochemistry in early-stage (stage 0 and stage I) breast cancers revealed high expression in low-grade lesions. Within a second independent early-stage non-high-grade cohort, we observed an association between TBX3 level in the DCIS and size of the invasive focus. Additionally, there was a positive correlation between TBX3 and SLUG, and TBX3 and TWIST1 in the invasive carcinoma. Pathway analysis revealed altered expression of several proteases and their inhibitors, consistent with the ability to degrade basement membrane in vivo. These findings strongly suggest the involvement of TBX3 in the promotion of invasiveness and progression of early-stage pre-invasive breast cancer to invasive carcinoma through the low-grade molecular pathway. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Epithelial-Mesenchymal Transition , Snail Family Transcription Factors/metabolism , T-Box Domain Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Cell Movement , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Signal Transduction , Snail Family Transcription Factors/genetics , T-Box Domain Proteins/genetics , Up-Regulation
3.
BMC Cancer ; 16(1): 671, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27553211

ABSTRACT

BACKGROUND: TBX3 is a T-box transcription factor repressor that is elevated in metastatic breast cancer and is believed to promote malignancy of tumor cells, possibly by promoting cell survival and epithelial-mesenchymal transition. METHODS: The relative expression of TBX3 was assessed in the 21T cell lines which were derived from an individual patient and represent three distinct stages of breast cancer progression: 21PT, atypical ductal hyperplasia; 21NT, ductal carcinoma in situ; and 21MT-1, invasive mammary carcinoma. Two different isoforms of TBX3 (TBX3iso1 and TBX3iso2) were overexpressed to evaluate cell survival/colony forming ability, growth, and invasion in the ductal carcinoma in situ-like 21NT cell line using an in vitro Matrigel model of cancer progression. In addition, TBX3 expression was knocked down to evaluate the effects of downregulating TBX3 on the invasive mammary carcinoma-like 21MT-1 cell line. Finally, PCR array profiling was used to assess alterations in gene expression due to TBX3 overexpression in the 21NT cells. RESULTS: TBX3 is abundant in the invasive 21MT-1 cell line, while being minimally expressed in the non-invasive 21NT and 21PT cell lines. Overexpression of either TBX3iso1 or TBX3iso2 in 21NT cells resulted in increased cell survival/colony forming ability, growth vs. apoptosis and invasion in Matrigel. In contrast, short hairpin RNA-mediated knockdown of TBX3 in the 21MT-1 cells resulted in smaller colonies, with a more regular, less dispersed (less infiltrative) morphology. Array profiling of the 21NT TBX3 iso1 and iso2 transfectants showed that there are common alterations in expression of several genes involved in signal transduction, cell cycle control/cell survival, epithelial-mesenchymal transition and invasiveness. CONCLUSIONS: Overall, these results indicate that TBX3 (isoform 1 or 2) expression can promote progression in a model of early breast cancer by altering cell properties involved in cell survival/colony formation and invasiveness, as well as key regulatory and EMT/invasiveness-related gene expressions.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Gene Expression Regulation, Neoplastic , Hyperplasia/pathology , T-Box Domain Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Collagen , Disease Progression , Drug Combinations , Epithelial-Mesenchymal Transition , Female , Humans , Hyperplasia/genetics , Hyperplasia/metabolism , Laminin , Neoplasm Invasiveness , Protein Isoforms , Proteoglycans , RNA, Small Interfering/genetics , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , Tumor Cells, Cultured
4.
Nitric Oxide ; 49: 26-39, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26068241

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is characterized by Von Hippel-Lindau (VHL)-deficiency, resulting in pseudohypoxic, angiogenic and glycolytic tumours. Hydrogen sulfide (H2S) is an endogenously-produced gasotransmitter that accumulates under hypoxia and has been shown to be pro-angiogenic and cytoprotective in cancer. It was hypothesized that H2S levels are elevated in VHL-deficient ccRCC, contributing to survival, metabolism and angiogenesis. Using the H2S-specific probe MeRhoAz, it was found that H2S levels were higher in VHL-deficient ccRCC cell lines compared to cells with wild-type VHL. Inhibition of H2S-producing enzymes could reduce the proliferation, metabolism and survival of ccRCC cell lines, as determined by live-cell imaging, XTT/ATP assay, and flow cytometry respectively. Using the chorioallantoic membrane angiogenesis model, it was found that systemic inhibition of endogenous H2S production was able to decrease vascularization of VHL-deficient ccRCC xenografts. Endogenous H2S production is an attractive new target in ccRCC due to its involvement in multiple aspects of disease.


Subject(s)
Carcinoma, Renal Cell/metabolism , Hydrogen Sulfide/antagonists & inhibitors , Hydrogen Sulfide/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chick Embryo , Humans , Hydrogen Sulfide/pharmacology , Neovascularization, Pathologic/metabolism , Xenograft Model Antitumor Assays
5.
Breast Cancer Res ; 16(1): R8, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24451146

ABSTRACT

INTRODUCTION: Osteopontin (OPN) is a malignancy-associated glycoprotein that contributes functionally to tumor aggressiveness. In metastatic breast cancer, we previously demonstrated that elevated OPN in primary tumor and blood was associated with poor prognosis. METHODS: We measured OPN in plasma by ELISA, and in tumors by immunohistochemistry, in 624 (94%) and 462 (69%), respectively, of 667 postmenopausal women with hormone responsive early breast cancer treated by surgery followed by adjuvant treatment with tamoxifen +/- octreotide in a randomized trial (NCIC CTG MA.14; National Cancer Institute of Canada Clinical Trials Group Mammary.14). RESULTS: Plasma OPN was measured in 2,540 samples; 688 at baseline and 1,852 collected during follow-up. Mean baseline plasma OPN was 46 ng/ml (range 22.6 to 290) which did not differ from normal levels. Mean percentage OPN tumor cell positivity was 33.9 (95% CI: 30.2 to 37.9). There was no correlation between plasma and tumor OPN values. In multivariate analysis, neither was associated with event-free survival (EFS), relapse-free survival (RFS), overall survival (OS), bone RFS or non-bone RFS. An exploratory analysis in patients with recurrence showed higher mean OPN plasma levels 60.7 ng/ml (23.9 to 543) in the recurrence period compared with baseline levels. CONCLUSIONS: The hypothesis that OPN tumor expression would have independent prognostic value in early breast cancer was not supported by multivariate analysis of this study population. Plasma OPN levels in women with hormone responsive early breast cancer in the MA.14 trial were not elevated and there was no evidence for prognostic value of plasma OPN in this defined group of patients. However, our finding of elevated mean OPN plasma level around the time of recurrence warrants further study. TRIAL REGISTRATION: NCT00002864, http://clinicaltrials.gov/show/NCT00002864.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/blood , Neoplasm Recurrence, Local/blood , Osteopontin/blood , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Disease-Free Survival , Female , Humans , Middle Aged , Octreotide/therapeutic use , Tamoxifen/therapeutic use
6.
Lab Invest ; 91(8): 1181-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21502940

ABSTRACT

Maspin (mammary serine protease inhibitor or SerpinB5) acts as a tumor suppressor when overexpressed in aggressive cancer cell lines. However, its role in human cancer is controversial. Maspin expression has been associated with a poor prognosis in some studies, whereas in others, with favorable outcome. The clinical data suggest, however, that nuclear-localized maspin is associated with improved survival. We hypothesized that the tumor suppressor activity of maspin may require nuclear localization, and that the discordance between clinical and experimental reports is a consequence of the variable subcellular distribution of maspin. Furthermore, we surmized that nuclear maspin could function as a tumor suppressor through the regulation of genes involved in tumor growth and invasion. Maspin or maspin fused to a nuclear export signal were expressed in metastatic human breast and epidermoid carcinoma cell lines. We found that pan-cellular localized maspin inhibited in vivo tumor growth and metastasis when assessed in xenograft chicken embryo and murine mammary fat pad injection models. However, when maspin was excluded from the nucleus via a nuclear exclusion signal, it no longer functioned as a metastasis suppressor. Using chromatin immunoprecipitation, we show that nuclear maspin was enriched at the promoter of colony-stimulating factor-1 (CSF-1) and associated with diminished levels of CSF-1 mRNA. Our findings demonstrate that the nuclear localization of maspin is required for its tumor and metastasis suppressor functions in vivo, and suggest that its mechanism of action involves, in part, direct association of maspin with target genes.


Subject(s)
Cell Nucleus/metabolism , Neoplasm Metastasis , Serpins/metabolism , Animals , Breast Neoplasms/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Chromatin/metabolism , Female , Humans , Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Nude , Promoter Regions, Genetic
7.
BMC Cancer ; 11: 25, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21247495

ABSTRACT

BACKGROUND: Osteopontin (OPN) is a secreted phosphoprotein often overexpressed at high levels in the blood and primary tumors of breast cancer patients. OPN contains two integrin-binding sites and a thrombin cleavage domain located in close proximity to each other. METHODS: To study the role of the thrombin cleavage site of OPN, MDA-MB-468 human breast cancer cells were stably transfected with either wildtype OPN (468-OPN), mutant OPN lacking the thrombin cleavage domain (468-ΔTC) or an empty vector (468-CON) and assessed for in vitro and in vivo functional differences in malignant/metastatic behavior. RESULTS: All three cell lines were found to equivalently express thrombin, tissue factor, CD44, αvß5 integrin and ß1 integrin. Relative to 468-OPN and 468-CON cells, 468-ΔTC cells expressing OPN with a deleted thrombin cleavage domain demonstrated decreased cell adhesion (p < 0.001), decreased mRNA expression of MCAM, maspin and TRAIL (p < 0.01), and increased uPA expression and activity (p < 0.01) in vitro. Furthermore, injection of 468-ΔTC cells into the mammary fat pad of nude mice resulted in decreased primary tumor latency time (p < 0.01) and increased primary tumor growth and lymph node metastatic burden (p < 0.001) compared to 468-OPN and 468-CON cells. CONCLUSIONS: The results presented here suggest that expression of thrombin-uncleavable OPN imparts an early tumor formation advantage as well as a metastatic advantage for breast cancer cells, possibly due to increased proteolytic activity and decreased adhesion and apoptosis. Clarification of the mechanisms responsible for these observations and the translation of this knowledge into the clinic could ultimately provide new therapeutic opportunities for combating breast cancer.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Osteopontin/genetics , Sequence Deletion , Thrombin/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Integrins/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Osteopontin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serpins/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Thromboplastin/metabolism , Transfection , Transplantation, Heterologous
8.
Lab Invest ; 90(8): 1247-58, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20458274

ABSTRACT

Early breast cancer progression involves advancement through specific morphological stages including atypical ductal hyperplasia (ADH), ductal carcinoma in situ (DCIS) and invasive mammary carcinoma (IMC), although not necessarily always in a linear fashion. Observational studies have examined genetic, epigenetic and gene expression differences in breast tissues representing these stages of progression, but model systems which would allow for experimental testing of specific factors influencing transition through these stages are scarce. The 21T series cell lines, all originally derived from the same patient with metastatic breast cancer, have been proposed to represent a mammary tumor progression series. We report here that three of the 21T cell lines indeed mimic specific stages of human breast cancer progression (21PT-derived cells, ADH; 21NT-derived cells, DCIS; 21MT-1 cells, IMC) when grown in the mammary fat pad of nude mice, albeit after a year. To develop a more rapid, readily manipulatable in vitro assay for examining the biological differences between these cell lines, we have used a 3D Matrigel system. When the three cell lines were grown in 3D Matrigel, they showed characteristic morphologies, in which quantifiable aspects of stage-specific in vivo behaviors (ie, differences in acinar structure formation, cell polarization, colony morphology, cell proliferation, cell invasion) were recapitulated in a reproducible fashion. Gene expression profiling revealed a characteristic pattern for each of the three cell lines. Interestingly, Wnt pathway alterations are particularly predominant in the early transition from 21PTci (ADH) to 21NTci (DCIS), whereas alterations in expression of genes associated with control of cell motility and invasion phenomena are more prominent in the later transition of 21NTci (DCIS) to 21MT-1 (IMC). This system thus reveals potential therapeutic targets and will provide a means of testing the influences of identified genes on transitions between these stages of pre-malignant to malignant growth.


Subject(s)
Breast Neoplasms , Breast/metabolism , Carcinoma in Situ/pathology , Carcinoma, Ductal/pathology , Carcinoma/pathology , Animals , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Carcinoma, Ductal/genetics , Carcinoma, Ductal/metabolism , Collagen , Disease Progression , Drug Combinations , Female , Gene Expression , Gene Expression Profiling , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Laminin , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Processes , Proteoglycans
9.
Cytometry A ; 75(4): 344-55, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18855920

ABSTRACT

The inability to sensitively detect metastatic cells in preclinical models of cancer has created challenges for studying metastasis in experimental systems. We previously developed a flow cytometry (FCM) method for quantifying circulating tumor cells (CTCs) in mouse models of breast cancer. We have adapted this methodology for analysis of tumor dissemination to bone marrow (BM) and lymph node (LN), and for analysis of these samples by laser scanning cytometry (LSC). Our objective was to implement these methodologies for characterization of tumor cell dissemination in preclinical models of cancer metastasis. Human cancer cells were injected into mice via mammary fat pad (MFP; spontaneous metastasis), tail vein (TV; targets lung), or intracardiac (IC; targets bone) routes. At several time points postinjection (4 h to 8 weeks), mice were sacrificed and blood, LNs, and BM were collected. Samples were immunomagnetically enriched and labeled with human leukocytic antigen-fluorescein isothiocyanate and CD45-PE antibodies (FCM/LSC), and propidium iodide (FCM) prior to quantitative analysis. Following MFP injection, CTCs increased over time, as did disseminated cells to the LN. Interestingly, tumor cells also spontaneously disseminated to BM, peaking at 2 weeks postinjection. Following TV injection, CTCs were initially high but decreased rapidly by 1 week before increasing to peak at endpoint. Combined with an observed concurrent increase in disseminated cells to LN and BM, this suggests that tumor cells may shed into the circulation from lung metastases that establish following initial cell delivery. Following IC injection, CTCs increased over time, peaking at 4 weeks. Tumor cells in the BM (most prevalent site of metastasis after IC injection) remained at moderate levels until peaking at endpoint. Combined use of FCM and LSC allows sensitive quantification of disseminated tumor cells in preclinical models of metastasis. These methods will be valuable for future studies aimed at testing new therapeutics in the metastatic setting.


Subject(s)
Flow Cytometry/methods , Laser Scanning Cytometry/methods , Neoplasm Metastasis/physiopathology , Neoplasms/physiopathology , Animals , Antibodies , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Flow Cytometry/instrumentation , Fluorescein-5-isothiocyanate , HLA Antigens/analysis , HLA Antigens/genetics , HLA Antigens/metabolism , Humans , Laser Scanning Cytometry/instrumentation , Leukocyte Common Antigens/analysis , Mice , Mice, Nude , Neoplasm Metastasis/pathology , Neoplasms/pathology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/physiopathology , Time Factors
10.
Int J Cancer ; 123(3): 526-34, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18470911

ABSTRACT

Breast cancer metastasis suppressor 1 (BRMS1) inhibits the ability of multiple human and murine cancer cell lines to metastasize to lymph nodes, bones and lungs. Comparison of mRNA expression in metastatic MDA-MB-435 human carcinoma cells (435) and metastasis-suppressed BRMS1 transfectants (435/BRMS1) showed a marked (>90%) reduction of osteopontin (OPN) mRNA and protein expression in BRMS1-overexpressing cells. OPN expression is associated with disease progression in patients, with higher levels of OPN produced by cancer cells associated with poorer patient survival. Furthermore, OPN has been suggested to promote survival of cancer cells in response to stress, although the mechanisms by which this may occur remain poorly understood. This study tested the hypothesis that re-expression of OPN in metastasis-suppressed 435/BRMS1 cells would reverse metastasis suppression and confer protection from stress-induced apoptosis. A stable pooled population of OPN overexpressing 435/BRMS1 cells was created (435/BRMS1/OPN). OPN re-expression did not affect in vitro cell growth rates; however, increased anchorage independent growth/survival and protection from hypoxia-induced apoptosis was observed (p < 0.05). In vivo, OPN re-expression in BRMS1 transfected cells did not affect in vivo primary tumor growth but did increase the incidence of spontaneous metastasis to lymph nodes and lungs in mice. These novel findings suggest that OPN downregulation by BRMS1 may be responsible, at least in part, for BRMS1-mediated metastasis suppression by sensitizing cancer cells to stress induced apoptosis. These studies clarify one mechanism by which BRMS1 can suppress metastasis.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neoplasm Proteins/metabolism , Osteopontin/metabolism , Animals , Apoptosis , Blotting, Northern , Blotting, Western , Cell Proliferation , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Repressor Proteins
11.
Clin Exp Metastasis ; 25(7): 727-40, 2008.
Article in English | MEDLINE | ID: mdl-18543067

ABSTRACT

The majority of breast cancer related deaths occur as a result of metastasis. The failure of effective treatments for metastasis is the underlying cause for this. Much remains unknown about the process of metastasis and how best to prevent or treat metastatic breast cancer. Therefore, a better understanding of the metastatic process is needed in order to determine effective therapeutic interventions to either eradicate, or slow down metastatic outgrowth of breast cancer. Metastasis is an inefficient process, however the ability of only a small number of cells to complete this process may have serious, life-threatening consequences. Little is known about whether expression of the metastasis suppressor breast cancer metastasis suppressor 1 (BRMS1) can suppress metastatic outgrowth in different organs in multiple experimental models of metastasis, or what effect BRMS1 expression has on the various steps in metastatic cascade. In this study we investigated the effect of BRMS1 expression on organ-specific metastasis. In addition, the steps in metastasis that are inhibited by BRMS1-expression were determined. In vivo, BRMS1 expression reduced metastatic burden to liver, bone, brain, and lung in mice by at least 75% (P<0.05). Detailed quantitative analysis of the metastatic process in lung showed that BRMS1 expression significantly reduced the numbers of solitary single cells that survive after initial arrest within the lung microvasculature, and also inhibited the initiation of growth subsequent to arrest. In vitro, BRMS1 expression decreased cancer cell survival under stress conditions (hypoxia), increased anoikis, and decreased the ability of cancer cells to adhere. These novel findings demonstrate that BRMS1 is a potent suppressor of metastasis in multiple organs, and identify two steps in the metastatic process that are sensitive to inhibition by BRMS1.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/physiology , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease Models, Animal , Female , Humans , Mice , Repressor Proteins
12.
Breast Cancer Res Treat ; 112(2): 243-54, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18097747

ABSTRACT

Osteopontin (OPN) has been clinically and experimentally associated with breast cancer metastasis. Proteolytic cleavage of OPN by thrombin has been reported to increase its biologic activity. The purpose of this study was to determine if inhibition of thrombin could reduce the malignancy-promoting effects of OPN on breast cancer cell behavior in vitro and in vivo. MDA-MB-468 human breast cancer cells were stably transfected to overexpress OPN (468-OPN) or a control vector (468-CON) and compared for functional differences in malignant/metastatic behavior in response to treatment with the thrombin-specific inhibitor Argatroban. Western blot analysis revealed that both 468-CON and 468-OPN cells produce thrombin and the thrombin-related protein tissue factor, and express very low levels of thrombin receptor (PAR-1). In vitro assays demonstrated that Argatroban treatment (25 microg/ml) of 468-OPN cells resulted in decreased cell growth, colony-forming ability, adhesion, and migration relative to untreated controls (P < 0.05), but did not have a significant effect on 468-CON cells. Following mammary fat pad injection, treatment with Argatroban (9 mg/kg/day) increased the in vivo tumor latency of both 468-CON and 468-OPN cells, and reduced primary tumor growth of 468-OPN cells (relative to untreated controls; P < 0.05). Furthermore, Argatroban treatment significantly decreased lymphatic metastasis of both 468-CON (P < 0.04) and 468-OPN (P < 0.01) cells relative to untreated controls. These novel findings indicate that inhibition of thrombin can reduce malignant and metastatic behavior of MDA-MB-468 breast cancer cells using both OPN-dependent and OPN-independent mechanisms, and suggest that thrombin inhibitors such as Argatroban may hold potential as therapeutic agents to combat breast cancer progression.


Subject(s)
Breast Neoplasms/drug therapy , Mammary Neoplasms, Animal/drug therapy , Osteopontin/physiology , Pipecolic Acids/pharmacology , Animals , Anticoagulants/pharmacology , Arginine/analogs & derivatives , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Female , Humans , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Metastasis , Osteopontin/metabolism , Platelet Aggregation Inhibitors/pharmacology , Receptors, Thrombin/metabolism , Sulfonamides , Thrombin/antagonists & inhibitors , Thrombin/chemistry
13.
Invest Radiol ; 43(2): 92-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18197061

ABSTRACT

OBJECTIVES: To determine a timepoint after contrast injection that yields equal liver parenchymal and vascular enhancement in micro-computed tomography images. To evaluate the utility of images acquired during this time period for the noninvasive measurement of liver-tumor volume. MATERIALS AND METHODS: The imaging timepoint was determined by quantifying the enhancement kinetics of Fenestra VC (0.015 mL/g) in NIH III mice. In respiratory-gated images of tumor bearing mice, the ability to measure tumor volume was evaluated with a measurement variability study, and by comparing in vivo and histologically measured tumor volume. RESULTS: Eight hours after contrast injection the liver parenchyma and vasculature were equally enhanced allowing for clear delineation of the unenhanced tumors. The smallest tumor detected in this study was 1.1 mm in diameter. The coefficient of variation for tumor-volume measurement ranged from 3.6% to 12.9% and from 6.3% to 25.8% for intra and interobserver variability, respectively. In vivo and histologic tumor-volume measurements were closely correlated (r = 0.98, P < 0.0001). CONCLUSIONS: Imaging at a time period of equal liver parenchyma and vascular enhancement after contrast injection allows for clear delineation of liver-tumor borders, thereby enabling quantitative tumor-volume monitoring.


Subject(s)
Contrast Media , Liver Neoplasms/diagnosis , Liver/pathology , Tomography, X-Ray Computed , Animals , Female , Image Processing, Computer-Assisted , Iopamidol , Liver Diseases/diagnosis , Liver Diseases/pathology , Liver Neoplasms/pathology , Mice , Models, Biological , Time Factors , Tumor Burden
14.
Cancer Res ; 65(8): 3396-403, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15833874

ABSTRACT

Metastatic spread, not primary tumor burden, is the leading cause of breast cancer deaths. For patient prognosis to improve, new systemic adjuvant therapies that are capable of effectively inhibiting the outgrowth of seeded tumor cells after surgical treatment of the primary breast tumor are needed. To facilitate the preclinical development of such therapies, relevant animal models of breast cancer metastasis that can mimic the postsurgical adjuvant setting are required. Here we developed a preclinical xenograft model of breast cancer metastasis where the primary tumor was removed by surgical resection before systemic adjuvant treatment. We used this model to assess the antimetastatic effect of postsurgical dietary intervention with the soy isoflavone genistein. The anticancer activity of genistein has been established in vitro and in vivo, however, few studies have tested the potential of genistein as an antimetastatic therapy. Using our model, we tested the efficacy of adjuvant treatment with genistein to inhibit the outgrowth of metastases postsurgery. To establish primary tumors, human breast carcinoma cells, MDA-MB-435/HAL, were implanted into the mammary fat pad of female nude mice. Primary tumors were left to grow for 5 weeks before being surgically removed. Mice were then randomized into two diet groups: control soy-free diet versus genistein-supplemented diet. Five weeks later, metastatic burden was assessed. Genistein reduced the percent metastatic burden in the lungs by 10-fold. These results indicate that dietary intervention following cancer surgery can affect the outgrowth of seeded tumor cells. The availability of well-characterized, clinically relevant animal models for studying factors that regulate metastatic outgrowth postsurgery will provide an important tool for developing new systemic adjuvant therapies.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/prevention & control , Genistein/administration & dosage , Animals , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Cell Growth Processes/drug effects , Cell Line, Tumor , Dietary Supplements , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Mice , Mice, Nude , Neoplasm Metastasis , Xenograft Model Antitumor Assays
15.
Cancer Res ; 65(12): 5231-7, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958568

ABSTRACT

Liver metastasis is a clinically significant contributor to the mortality associated with melanoma, colon, and breast cancer. Preclinical mouse models are essential to the study of liver metastasis, yet their utility has been limited by the inability to study this dynamic process in a noninvasive and longitudinal manner. This study shows that three-dimensional high-frequency ultrasound can be used to noninvasively track the growth of liver metastases and evaluate potential chemotherapeutics in experimental liver metastasis models. Liver metastases produced by mesenteric vein injection of B16F1 (murine melanoma), PAP2 (murine H-ras-transformed fibroblast), HT-29 (human colon carcinoma), and MDA-MB-435/HAL (human breast carcinoma) cells were identified and tracked longitudinally. Tumor size and location were verified by histologic evaluation. Tumor volumes were calculated from the three-dimensional volumetric data, with individual liver metastases showing exponential growth. The importance of volumetric imaging to reduce uncertainty in tumor volume measurement was shown by comparing three-dimensional segmented volumes with volumes estimated from diameter measurements and the assumption of an ellipsoid shape. The utility of high-frequency ultrasound imaging in the evaluation of therapeutic interventions was established with a doxorubicin treatment trial. These results show that three-dimensional high-frequency ultrasound imaging may be particularly well suited for the quantitative assessment of metastatic progression and the evaluation of chemotherapeutics in preclinical liver metastasis models.


Subject(s)
Liver Neoplasms, Experimental/diagnostic imaging , Liver Neoplasms, Experimental/secondary , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Disease Models, Animal , HT29 Cells , Humans , Liver Neoplasms, Experimental/pathology , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Mice, SCID , Necrosis , Ultrasonography
16.
Cancer Res ; 63(16): 4763-5, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12941790

ABSTRACT

Some clinical studies suggest that timing of surgery during specific menstrual phases may influence the chances of survival for premenopausal women with breast cancer, whereas other studies failed to find this effect. Because most breast cancer deaths are attributable to metastases, we hypothesized that aspects of the metastatic process might be sensitive to cyclic hormonal fluctuations. Our goal was to develop a mouse model to assess possible mechanisms for the effect of the menstrual cycle on metastatic ability. To separate the effects of the hormonal milieu on the host versus the cancer cells, we began by using melanoma cells. We report here that the estrous phase at the time of entry of B16F10 melanoma cells into the circulation leads to marked differences in organ-specific metastasis, suggesting that this concept merits additional study.


Subject(s)
Estrus/physiology , Melanoma, Experimental/secondary , Animals , Female , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Organ Specificity , Ovarian Neoplasms/secondary
17.
Cancer Biol Ther ; 4(12): 1336-41, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16258259

ABSTRACT

There is a need for molecular markers that predict biological behavior of adult soft tissue tumors. Elevated levels of osteopontin (OPN) a transformation-linked protein, have been associated with poor survival in many cancers. OPN induces cell migration in cancer cells, in part through activation of the hepatocyte growth factor (HGF) receptor (Met) and its signaling pathway. Met expression has been associated with a poor prognosis in some sarcomas. In a series of 15 patients with adult soft tissue tumors, we found that mRNA levels of OPN (p=0.015), Met (p=0.03) and HGF (p<0.001) were significantly higher in tumor tissues relative to paired normal tissues. By immunohistochemistry, in tumor tissue from 33 patients, we demonstrated that increased expression of OPN, but not Met protein, was associated with higher stage (p=0.025) and grade (p=0.005). We found that increased expression of OPN, but not Met protein, was associated with decreased overall survival (p=0.008) at five years. This study, which is the first to examine coexpression of these two markers, suggests that OPN may have potential as a prognostic marker in adult soft tissue sarcomas, and further that OPN+/-Met signaling pathways may contribute to their biological behavior.


Subject(s)
Gene Expression , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/metabolism , Sialoglycoproteins/metabolism , Soft Tissue Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Female , Hepatocyte Growth Factor/genetics , Humans , Male , Middle Aged , Osteopontin , Proto-Oncogene Proteins c-met/genetics , Sialoglycoproteins/genetics , Soft Tissue Neoplasms/pathology
18.
Clin Exp Metastasis ; 22(4): 351-61, 2005.
Article in English | MEDLINE | ID: mdl-16170671

ABSTRACT

Breast cancer often spreads from the primary tumor to regional lymph nodes. Lymph node status provides clinically important information for making treatment decisions. Spread via lymphatics is also important for the biology of breast cancer, as tumor cells in lymph nodes may provide a reservoir of cells leading to distant, lethal metastases. Improved understanding of the biology of lymphatic spread thus is important for improved breast cancer survival. Advances towards understanding the interactions between tumors cells and lymphatic vessels have in part been limited by the lack of suitable cell lines and experimental models. We have addressed this need by developing a new model of lymphatic metastasis. Here we describe the establishment of 468LN cells, a variant of the MDA-MB-468 human breast adenocarcinoma cell line, which produces extensive lymph node metastasis following orthotopic injection of nude mice. 468LN cells are also more aggressive in vitro, produce more osteopontin and express different surface integrins compared to the parent line. The dramatic in vitro and in vivo phenotypic and molecular differences of 468LN and parental 468GFP cells make this pair of cell lines a unique model for the specific study of lymph node metastasis of breast cancer.


Subject(s)
Adenocarcinoma/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Lymphatic Metastasis/pathology , Mice , Animals , Female , Humans , Lymph Nodes/pathology , Mice, Nude , Neoplasm Transplantation
19.
Mol Cancer Res ; 1(11): 810-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14517343

ABSTRACT

Osteopontin (OPN) is a secreted phosphoprotein that has been associated with malignancy of breast and other cancers. OPN binds to several cell surface integrins including alpha(v)beta(3), alpha(v)beta(5), and alpha(v)beta(1). Although the relative contribution of these integrins to breast cancer cell malignancy is uncertain, correlative studies suggest that alpha(v)beta(3) may be particularly associated with increased tumor aggressiveness. Previously, we reported that tumorigenic, nonmetastatic 21NT mammary carcinoma cells respond to OPN through alpha(v)beta(5) and alpha(v)beta(1) but not alpha(v)beta(3). Here, we determined that 21NT cells lack beta(3) expression, and we asked whether expression of alpha(v)beta(3) could enhance the ability of breast cancer cells to respond to the malignancy-promoting effects of OPN both in vitro and in vivo. 21NT cells stably transfected with beta(3) showed significantly increased adhesion, migration, and invasion to OPN in vitro compared with vector control. To determine if beta(3) could also enhance the response of breast epithelial cells to OPN in vivo, cells stably transfected with both beta(3) and OPN (NT/Obeta(3)) were injected into the mammary fat pad of female nude mice and primary tumor growth was assessed relative to controls. Mice injected with NT/Obeta(3) cells demonstrated a significantly increased primary tumor take (75% of mice) compared with controls (0-12.5% of mice) as well as a decreased tumor doubling time and a decreased tumor latency period. These results suggest that increased expression of the alpha(v)beta(3) integrin during breast cancer progression can make tumor cells more responsive to malignancy-promoting ligands such as OPN and result in increased tumor cell aggressiveness.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Integrin beta3/genetics , Integrin beta3/metabolism , Sialoglycoproteins/metabolism , Sialoglycoproteins/pharmacology , Animals , Blotting, Northern , Breast Neoplasms/genetics , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Female , Flow Cytometry , Humans , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Osteopontin , Sialoglycoproteins/genetics
20.
Cell Rep ; 8(5): 1558-70, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25176655

ABSTRACT

Tumor cell extravasation is a key step during cancer metastasis, yet the precise mechanisms that regulate this dynamic process are unclear. We utilized a high-resolution time-lapse intravital imaging approach to visualize the dynamics of cancer cell extravasation in vivo. During intravascular migration, cancer cells form protrusive structures identified as invadopodia by their enrichment of MT1-MMP, cortactin, Tks4, and importantly Tks5, which localizes exclusively to invadopodia. Cancer cells extend invadopodia through the endothelium into the extravascular stroma prior to their extravasation at endothelial junctions. Genetic or pharmacological inhibition of invadopodia initiation (cortactin), maturation (Tks5), or function (Tks4) resulted in an abrogation of cancer cell extravasation and metastatic colony formation in an experimental mouse lung metastasis model. This provides direct evidence of a functional role for invadopodia during cancer cell extravasation and distant metastasis and reveals an opportunity for therapeutic intervention in this clinically important process.


Subject(s)
Cell Surface Extensions/metabolism , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Transcellular Cell Migration , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/pharmacology , Benzodioxoles/pharmacology , Cell Line, Tumor , Cell Surface Extensions/drug effects , Chick Embryo , Cortactin/genetics , Cortactin/metabolism , Female , Humans , Lung Neoplasms/pathology , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Nude , Neoplasm Metastasis , Neoplastic Stem Cells/physiology , Phosphate-Binding Proteins , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL