Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 15(4): e0231762, 2020.
Article in English | MEDLINE | ID: mdl-32348319

ABSTRACT

Canine osteosarcoma (OSA), the most common canine primary bone malignancy, has a highly aggressive biologic behavior. Despite current standard of care therapies, including amputation and adjuvant chemotherapy, most dogs still succumb to metastatic disease. Further investigations into molecular mechanisms and pathways driving OSA are needed to improve therapeutic options. The Hedgehog (HH) cell-signaling pathway has demonstrated involvement in human OSA. Several studies in canine OSA have found changes in expression of some HH pathway genes and demonstrated a role for HH transcription factors. However, the role of this pathway as well as the translational value of its targeting in canine OSA are still undefined. The objectives of this study were to determine the expression of HH components directly in canine OSA tissues and to evaluate the biologic impact of HH signaling inhibition in canine OSA cells. In situ hybridization was used to detect HH family mRNA expression in archived canine OSA tissues and revealed variable expression levels of these mRNAs in canine OSA tissues. The effect of a commercially available Smoothened inhibitor, vismodegib, was studied in established canine OSA cell lines. Alterations in cellular growth as well as assessment of downstream HH targets were evaluated. Although changes in cell growth were noted following Smoothened inhibition, inconsistent decreases in target gene expression were found. While treatment with vismodegib had a negative impact on canine OSA cell growth and viability, the mechanism remains unclear. Further studies are warranted to evaluate the clinical significance of canonical HH signaling in canine OSA.


Subject(s)
Anilides/pharmacology , Bone Neoplasms/pathology , Hedgehog Proteins/metabolism , Osteosarcoma/pathology , Pyridines/pharmacology , Signal Transduction/drug effects , Anilides/therapeutic use , Animals , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/veterinary , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dogs , Gene Expression Profiling , Hedgehog Proteins/genetics , Osteosarcoma/drug therapy , Osteosarcoma/veterinary , Pyridines/therapeutic use , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism
2.
J Pathol ; 218(3): 399-408, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19402129

ABSTRACT

The Met receptor tyrosine kinase (RTK) is aberrantly expressed in human osteosarcoma and is an attractive molecular target for cancer therapy. We studied spontaneous canine osteosarcoma (OSA) as a potential pre-clinical model for evaluation of Met-targeted therapies. The canine MET oncogene exhibits 90% homology compared with human MET, indicating that cross-species functional studies are a viable strategy. Expression and activation of the canine Met receptor were studied utilizing immunohistochemical techniques in 39 samples of canine osteosarcoma, including 35 primary tumours and four metastases. Although the Met RTK is barely detectable in primary culture of canine osteoblasts, high expression of Met protein was observed in 80% of canine osteosarcoma samples acquired from various breeds. Met protein overexpression was also concordant with its activation as indicated by phosphorylation of critical tyrosine residues. In addition, Met was expressed and constitutively activated in canine osteosarcoma cell lines. OSA cells expressing high levels of Met demonstrated activation of downstream transducers, elevated spontaneous motility, and invasiveness which were impaired by both a small molecule inhibitor of Met catalytic activity (PHA-665752) and met-specific, stable RNA interference obtained by means of lentiviral vector. Similar to observations in human OSA, these data suggest that Met is commonly overexpressed and activated in canine OSA and that inhibition of Met impairs the invasive and motogenic properties of canine OSA cells. These data implicate Met as a potentially important factor for canine OSA progression and indicate that it represents a viable model to study Met-targeted therapies.


Subject(s)
Bone Neoplasms/metabolism , Disease Models, Animal , Osteosarcoma/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Antineoplastic Agents/pharmacology , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Enzyme Inhibitors/pharmacology , Humans , Indoles/pharmacology , Neoplasm Invasiveness , Osteosarcoma/pathology , Osteosarcoma/secondary , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , RNA Interference , Sulfones/pharmacology
3.
J Biol Chem ; 283(12): 7616-27, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18184652

ABSTRACT

Malignant breast cancer cells that have entered the blood circulation from primary mammary fat pad tumors or are grown in end-over-end suspension culture assemble a characteristic, multi-globular polymeric fibronectin (polyFn) coat on their surfaces. Surface polyFn is critical for pulmonary metastasis, presumably by facilitating lung vascular arrest via endothelial dipeptidylpeptidase IV (CD26). Here, we show that cell-surface polyFn assembly is initiated by the state of suspension, is dependent upon the synthesis and secretion of cellular Fn, and is augmented in a dose- and time-dependent manner by plasma Fn. PolyFn assembly is regulated by protein kinase Cepsilon (PKCepsilon), which translocates rapidly and in increasing amounts from the cytosol to the plasma membrane and is phosphorylated. PolyFn assembly is impeded by select inhibitors of this kinase, i.e. bisindolylmaleimide I, Ro-32-0432, Gö6983, and Rottlerin, by the phorbol 12-myristate 13-acetate-mediated and time-dependent loss of PKCepsilon protein and decreased plasma membrane translocation, and more specifically, by stable transfection of lung-metastatic MTF7L breast cancer cells with small interfering RNA-PKCepsilon and dominant-negative PKCepsilon constructs (e.g. RD-PKCepsilon). The inability to assemble a cell surface-associated polyFn coat by knockdown of endogenous Fn or PKCepsilon impedes cancer cells from metastasis to the lungs. The present studies identify a novel regulatory mechanism for polyFn assembly on blood-borne breast cancer cells and depict its effect on pulmonary metastasis.


Subject(s)
Fibronectins/metabolism , Lung Neoplasms/enzymology , Mammary Neoplasms, Animal/enzymology , Neoplasm Proteins/metabolism , Neoplastic Cells, Circulating/metabolism , Protein Kinase C-epsilon/metabolism , Animals , Cell Membrane/enzymology , Cell Membrane/metabolism , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Female , Fibronectins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/genetics , Neoplasm Proteins/genetics , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/genetics , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Protein Transport/genetics , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...