Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
BMC Cancer ; 15: 901, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26573603

ABSTRACT

BACKGROUND: AXL is a well-characterized, protumorigenic receptor tyrosine kinase that is highly expressed and activated in numerous human carcinomas and sarcomas, including aggressive subtypes of liposarcoma. However, the role of AXL in the pathogenesis of well-differentiated (WDLPS), dedifferentiated (DDLPS), and pleomorphic liposarcoma (PLS) has not yet been determined. METHODS: Immunohistochemical analysis of AXL expression was conducted on two tissue microarrays containing patient WDLPS, DDLPS, and PLS samples. A panel of DDLPS and PLS cell lines were interrogated via western blot for AXL expression and activity and by ELISA for growth arrest-specific 6 (GAS6) production. AXL knockdown was achieved by siRNA or shRNA. The effects of AXL knockdown on cell proliferation, migration, and invasion were measured in vitro. In addition, AXL shRNA-containing DDLPS cells were assessed for their tumor-forming capacity in vivo. RESULTS: In this study, we determined that AXL is expressed in a subset of WDLPS, DDLPS, and PLS patient tumor samples. In addition, AXL and its ligand GAS6 are expressed in a panel of DDLPS and PLS cell lines. We show that the in vitro activation of AXL via stimulation with exogenous GAS6 resulted in a significant increase in cell proliferation, migration, and invasion in DDLPS and PLS cell lines. Transient knockdown of AXL resulted in attenuation of these protumorigenic phenotypes in vitro. Stable AXL knockdown not only decreased migratory and invasive characteristics of DDLPS and PLS cells in vitro but also significantly diminished tumorigenicity of two dedifferentiated liposarcoma xenograft models in vivo. CONCLUSIONS: Our results suggest that AXL signaling contributes to the aggressiveness of DDLPS and PLS, and that AXL is therefore a potential therapeutic target for treatment of these rare, yet devastating tumors.


Subject(s)
Liposarcoma/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Liposarcoma/pathology , Neoplasm Invasiveness/physiopathology , Axl Receptor Tyrosine Kinase
3.
Cancer Res ; 72(7): 1751-62, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22350414

ABSTRACT

Liposarcoma can be an aggressive, debilitating, and fatal malignancy. In this study, we identifed miRNAs associated with the differentiation status of liposarcoma to gain insight into the basis for its progression. miRNA expression profiles determined in human tumors and normal fat specimens identified a dedifferentiated tumor expression signature consisting of 35 miRNAs. Deregulated miRNA expression was confirmed in a second independent sample cohort. The miR-155 was the most overexpressed miRNA and functional investigations assigned an important role in the growth of dedifferentiated liposarcoma cell lines. Transient or stable knockdown of miR-155 retarded tumor cell growth, decreased colony formation, and induced G(1)-S cell-cycle arrest in vitro and blocked tumor growth in murine xenografts in vivo. We identified casein kinase 1α (CK1α) as a direct target of miR-155 control which enhanced ß-catenin signaling and cyclin D1 expression, promoting tumor cell growth. In summary, our results point to important functions for miR-155 and ß-catenin signaling in progression of liposarcoma, revealing mechanistic vulnerabilities that might be exploited for both prognostic and therapeutic purposes.


Subject(s)
Casein Kinase I/genetics , Liposarcoma/genetics , MicroRNAs/physiology , Oncogenes , Signal Transduction/physiology , beta Catenin/physiology , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Cyclin D1/physiology , Gene Expression Regulation, Neoplastic , Humans , Liposarcoma/pathology , Mice , Molecular Sequence Data
4.
Clin Cancer Res ; 17(18): 5901-12, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21821699

ABSTRACT

PURPOSE: Molecular deregulations underlying epithelioid sarcoma (ES) progression are poorly understood yet critically needed to develop new therapies. Epidermal growth factor receptor (EGFR) is overexpressed in ES; using preclinical models, we examined the ES EGFR role and assessed anti-ES EGFR blockade effects, alone and with mTOR inhibition. EXPERIMENTAL DESIGN: EGFR and mTOR expression/activation was examined via tissue microarray (n = 27 human ES specimens; immunohistochemistry) and in human ES cell lines (Western blot and quantitative reverse transcriptase PCR). Cell proliferation, survival, migration, and invasion effects of EGFR and mTOR activation treated with erlotinib (anti-EGFR small-molecule inhibitor) alone and combined with rapamycin were assessed in cell culture assays. In vivo growth effects of erlotinib alone or with rapamycin were evaluated using severe combined immunodeficient mouse ES xenograft models. RESULTS: EGFR was expressed and activated in ES specimens and cell lines. EGFR activation increased ES cell proliferation, motility, and invasion and induced cyclin D1, matrix metalloproteinase (MMP) 2, and MMP9 expression. EGFR blockade inhibited these processes and caused significant cytostatic ES growth inhibition in vivo. mTOR pathway activation at varying levels was identified in all tissue microarray-evaluable ES tissues; 88% of samples had no or reduced PTEN expression. Similarly, both ES cell lines showed enhanced mTOR activity; VAESBJ cells exhibited constitutive mTOR activation uncoupled from EGFR signaling. Most importantly, combined erlotinib/rapamycin resulted in synergistic anti-ES effects in vitro and induced superior tumor growth inhibition in vivo versus single agent administration. CONCLUSIONS: EGFR and mTOR signaling pathways are deregulated in ES. Preclinical ES model-derived insights suggest that combined inhibition of these targets might be beneficial, supporting evaluations in clinical trials.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Sarcoma/drug therapy , Sarcoma/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Disease Models, Animal , Drug Synergism , ErbB Receptors/genetics , Erlotinib Hydrochloride , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Invasiveness/genetics , Quinazolines/pharmacology , Quinazolines/therapeutic use , Sarcoma/genetics , Sarcoma/pathology , Signal Transduction/drug effects , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/genetics , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 17(12): 3943-55, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21540237

ABSTRACT

PURPOSE: MET signaling has been suggested a potential role in malignant peripheral nerve sheath tumors (MPNST). Here, MET function and blockade were preclinically assessed. EXPERIMENTAL DESIGN: Expression levels of MET, its ligand hepatocyte growth factor (HGF), and phosphorylated MET (pMET) were examined in a clinically annotated MPNST tissue microarray (TMA) incorporating univariable and multivariable statistical analyses. Human MPNST cells were studied in vitro and in vivo; Western blot (WB) and ELISA were used to evaluate MET and HGF expression, activation, and downstream signaling. Cell culture assays tested the impact of HGF-induced MET activation and anti-MET-specific siRNA inhibition on cell proliferation, migration, and invasion; in vivo gel-foam assays were used to evaluate angiogenesis. Cells stably transduced with anti-MET short hairpin RNA (shRNA) constructs were tested for growth and metastasis in severe combined immunodeficient (SCID) mice. The effect of the tyrosine kinase inhibitor XL184 (Exelixis) targeting MET/VEGFR2 (vascular endothelial growth factor receptor 2) on local and metastatic MPNST growth was examined in vivo. RESULTS: All three markers were expressed in MPNST human samples; pMET expression was an independent prognosticator of poor patient outcome. Human MPNST cell lines expressed MET, HGF, and pMET. MET activation increased MPNST cell motility, invasion, angiogenesis, and induced matrix metalloproteinase-2 (MMP2) and VEGF expression; MET knockdown had inverse effects in vitro and markedly decreased local and metastatic growth in vivo. XL184 abrogated human MPNST xenograft growth and metastasis in SCID mice. CONCLUSIONS: Informative prognosticators and novel therapies are crucially needed to improve MPNST management and outcomes. We show an important role for MET in MPNST, supporting continued investigation of novel anti-MET therapies in this clinical context.


Subject(s)
Nerve Sheath Neoplasms/diagnosis , Proto-Oncogene Proteins c-met/metabolism , Anilides/pharmacology , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hepatocyte Growth Factor/metabolism , Humans , Mice , Mice, SCID , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/genetics , Nerve Sheath Neoplasms/mortality , Nerve Sheath Neoplasms/pathology , Phenotype , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/genetics , Pyridines/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Cancer Res ; 71(1): 185-96, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21084276

ABSTRACT

Histone deacetylase inhibitors (HDACi) show promise as cancer therapeutics; however, the full scope of their utility remains unknown. Here we report findings that strongly rationalize clinical evaluation of HDACis in malignant peripheral nerve sheath tumors (MPNST), a class of highly aggressive, therapeutically resistant, and commonly fatal malignancies that occur sporadically or in patients with the inherited neurofibromatosis type-1 (NF1) syndrome. We evaluated the effects of the chemical HDACis PCI-24781, suberoylanilide hydroxamic acid, and MS-275 on a panel of human NF1-associated and sporadic MPNSTs in vitro and in vivo. A subset of MPNSTs was found to be highly sensitive to HDACis, especially to PCI-24781. All cell lines in this group were NF1-associated. Significant proapoptotic effects were noted in vitro and in vivo and were independent of p53 mutational status. In contrast, as a group the sporadic-MPNST cells were markedly resistant to HDACi treatment. HDACis were found to induce productive autophagy in MPNST cells. Genetic and/or pharmacologic autophagy blockade resulted in significant HDACi-induced apoptosis in cells defined as resistant or sensitive, leading to abrogated growth of primary tumors and lung metastases in tumor xenograft assays. Among autophagy-associated genes expressed in response to HDACi, the immunity-related GTPase family, M was validated as a critical target in mediating HDACi-induced autophagy and enhanced apoptosis. Taken together, our findings strongly support the evaluation of HDACi currently in clinical trials as an important new therapeutic strategy to treat MPNST, including in combination with autophagy blocking combination regimens in particular for patients with sporadic MPNST.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Histone Deacetylase Inhibitors/pharmacology , Nerve Sheath Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Profiling , Histone Deacetylase Inhibitors/therapeutic use , Humans , Microscopy, Electron, Transmission , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/pathology
7.
Clin Cancer Res ; 15(10): 3472-83, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19417021

ABSTRACT

PURPOSE: Histone deactylase inhibitors (HDACi) are a promising new class of anticancer therapeutics; however, little is known about HDACi activity in soft tissue sarcoma (STS), a heterogeneous cohort of mesenchymal origin malignancies. Consequently, we investigated the novel HDACi PCI-24781, alone/in combination with conventional chemotherapy, to determine its potential anti-STS-related effects and the underlying mechanisms involved. EXPERIMENTAL DESIGN: Immunoblotting was used to evaluate the effects of PCI-24781 on histone and nonhistone protein acetylation and expression of potential downstream targets. Cell culture-based assays were utilized to assess the effects of PCI-24781 on STS cell growth, cell cycle progression, apoptosis, and chemosensitivity. Quantitative reverse transcription-PCR, chromatin immunoprecipitation, and reporter assays helped elucidate molecular mechanisms resulting in PCI-24781-induced Rad51 repression. The effect of PCI-24781, alone or with chemotherapy, on tumor and metastatic growth was tested in vivo using human STS xenograft models. RESULTS: PCI-24781 exhibited significant anti-STS proliferative activity in vitro, inducing S phase depletion, G(2)/M cell cycle arrest, and increasing apoptosis. Superior effects were seen when combined with chemotherapy. A PCI-24781-induced reduction in Rad51, a major mediator of DNA double-strand break homologous recombination repair, was shown and may be a mechanism underlying PCI-24781 chemosensitization. We showed that PCI-24781 transcriptionally represses Rad51 through an E2F binding-site on the Rad51 proximal promoter. Although single-agent PCI-24781 had modest effects on STS growth and metastasis, marked inhibition was observed when combined with chemotherapy. CONCLUSIONS: In light of these findings, this novel molecular-based combination may be applicable to multiple STS histologic subtypes, and potentially merits rigorous evaluation in human STS clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzofurans/therapeutic use , Histone Deacetylase Inhibitors , Hydroxamic Acids/therapeutic use , Sarcoma, Experimental/drug therapy , Animals , Apoptosis/drug effects , Benzofurans/administration & dosage , Benzofurans/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Exonucleases/genetics , Exonucleases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Immunohistochemistry , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Mice , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , S Phase/drug effects , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 6(5): 1650-60, 2007 May.
Article in English | MEDLINE | ID: mdl-17513613

ABSTRACT

We investigated whether Rad51 overexpression plays a role in soft tissue sarcoma (STS) chemoresistance as well as the regulatory mechanisms underlying its expression. The studies reported here show that Rad51 protein is overexpressed in a large panel of human STS specimens. Human STS cell lines showed increased Rad51 protein expression, as was also observed in nude rat STS xenografts. STS cells treated with doxorubicin exhibited up-regulation of Rad51 protein while arrested in the S-G(2) phase of the cell cycle. Treatment with anti-Rad51 small interfering RNA decreased Rad51 protein expression and increased chemosensitivity to doxorubicin. Because we previously showed that reintroduction of wild-type p53 (wtp53) into STS cells harboring a p53 mutation led to increased doxorubicin chemosensitivity, we hypothesized that p53 participates in regulating Rad51 expression in STS. Reintroduction of wtp53 into STS cell lines resulted in decreased Rad51 protein and mRNA expression. Using luciferase reporter assays, we showed that reconstitution of wtp53 function decreased Rad51 promoter activity. Deletion constructs identified a specific Rad51 promoter region containing a p53-responsive element but no p53 consensus binding site. Electrophoretic mobility shift assays verified activator protein 2 (AP2) binding to this region and increased AP2 binding to the promoter in the presence of wtp53. Mutating this AP2 binding site eliminated the wtp53 repressive effect. Furthermore, AP2 knockdown resulted in increased Rad51 expression. In light of the importance of Rad51 in modulating STS chemoresistance, these findings point to a potential novel strategy for molecular-based treatments that may be of relevance to patients burdened by STS.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Rad51 Recombinase/physiology , Sarcoma/drug therapy , Soft Tissue Neoplasms/drug therapy , Transcription Factor AP-2/physiology , Tumor Suppressor Protein p53/physiology , Cell Cycle , Cell Line, Tumor , Genes, Reporter , Humans , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , RNA, Small Interfering/metabolism , Rad51 Recombinase/metabolism , Response Elements
9.
Cancer Res ; 66(17): 8770-8, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951193

ABSTRACT

To better elucidate the role of vascular endothelial growth factor (VEGF)(165) in soft tissue sarcoma (STS) growth, metastasis, and chemoresistance, we generated stably transfected human STS cell lines with VEGF(165) to study the effect of VEGF(165) on STS cells in vitro and the effect of culture medium from these cells on human umbilical vascular endothelial cells. Severe combined immunodeficient mice bearing xenografts of transfected cell lines were used to assess the effect of VEGF overexpression and the effect of VEGF receptor (VEGFR) 2 inhibition on STS growth, metastasis, and response to doxorubicin. VEGF(165)-transfected xenografts formed highly vascular tumors with shorter latency, accelerated growth, enhanced chemoresistance, and increased incidence of pulmonary metastases. Blockade of VEGFR2 signaling using DC101 anti-VEGFR2 monoclonal antibody enhanced doxorubicin chemoresponse; this combined biochemotherapy inhibited tumor growth and decreased pulmonary metastases without overt toxicity. Combined therapy reduced microvessel counts while increasing vessel maturation index. VEGF overexpression did not affect on the sarcoma cells per se; however, conditioned medium from VEGF transfectants caused increased endothelial cell proliferation, migration, and chemoresistance. Addition of DC101 induced endothelial cell sensitivity to doxorubicin and suppressed the activity of matrix metalloproteinases secreted by endothelial cells. We therefore conclude that VEGF is a critical determinant of STS growth and metastasis and that STS chemoresistance, in our model, is a process induced by the interplay between STS cells and tumor-associated endothelial cells. STS growth and metastasis can be interrupted by combined low-dose doxorubicin and anti-VEGFR2, a strategy that attacks STS-associated endothelial cells. In the future, such therapeutic approaches may be useful in treating STS before the development of clinically apparent metastases.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Sarcoma/genetics , Sarcoma/pathology , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Division , Cell Line, Tumor , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Female , Humans , Mice , Mice, SCID , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Transfection , Transplantation, Heterologous , Umbilical Veins , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
10.
J Biol Chem ; 280(6): 4825-33, 2005 Feb 11.
Article in English | MEDLINE | ID: mdl-15563462

ABSTRACT

The protein kinase C (PKC) family consists of serine/threonine protein kinases that play important roles in signal transduction, cell proliferation, and tumor formation. Recent studies found that PKCs are commonly overexpressed in human tumors, including soft tissue sarcoma (STS). Overexpression of PKCs contributes to invasion and migration of tumor cells and induction of angiogenesis. PKC can also phosphorylate the multidrug resistance (MDR) gene-encoded P-glycoprotein and induce MDR phenotype. Our previous studies showed that mutation of p53 enhanced STS metastasis and mediated the MDR phenotype. Restoring wild type (WT) p53 in STS cells containing mutant p53 sensitized the cells to chemotherapy. In the present study, we found that PKCalpha protein expression is inhibited by WT p53 partly due to reduced PKCalpha mRNA expression in STS cells, but p53 does not affect PKCalpha mRNA stability. Deletion and mutation analysis of the PKCalpha promoter fused to the luciferase reporter gene identified a Sp1 binding site (-244/-234) in the PKCalpha promoter that is required for p53-mediated inhibition of PKCalpha promoter activity. More importantly, PKCalpha phosphorylates and activates MDR1 P-glycoprotein, whereas inhibition of PKCalpha by p53 leads to decreased MDR1 phosphorylation in STS cells, which sensitizes STS cells to chemotherapeutic agents. These data indicate that WT p53 may resensitize STS to chemotherapeutic agents by reducing MDR1 phosphorylation via transcriptional repression of PKCalpha expression. Thus, molecular-based therapies targeting mutant p53 and PKCalpha may be an effective new strategy to improve chemotherapeutic efficacy in STS.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Protein Kinase C/biosynthesis , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Binding Sites , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Dactinomycin/pharmacology , Down-Regulation , Gene Deletion , Genes, Reporter , Genes, p53 , Humans , Immunoprecipitation , Luciferases/metabolism , Models, Genetic , Mutation , Neovascularization, Pathologic , Phosphorylation , Point Mutation , Promoter Regions, Genetic , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-alpha , RNA/chemistry , RNA, Messenger/metabolism , Signal Transduction , Temperature , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...